US20080233637A1 - Melanoma antigens and their use in diagnostic and therapeutic methods - Google Patents

Melanoma antigens and their use in diagnostic and therapeutic methods Download PDF

Info

Publication number
US20080233637A1
US20080233637A1 US11/932,255 US93225507A US2008233637A1 US 20080233637 A1 US20080233637 A1 US 20080233637A1 US 93225507 A US93225507 A US 93225507A US 2008233637 A1 US2008233637 A1 US 2008233637A1
Authority
US
United States
Prior art keywords
seq
melanoma
mart
amino acid
hla
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US11/932,255
Other versions
US7803614B2 (en
Inventor
Yutaka Kawakami
Steven A. Rosenberg
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
US Government
Original Assignee
US Government
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=22869773&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20080233637(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from US09/073,138 external-priority patent/US6537560B1/en
Application filed by US Government filed Critical US Government
Priority to US11/932,255 priority Critical patent/US7803614B2/en
Publication of US20080233637A1 publication Critical patent/US20080233637A1/en
Priority to US12/851,729 priority patent/US8030280B2/en
Application granted granted Critical
Publication of US7803614B2 publication Critical patent/US7803614B2/en
Priority to US13/192,804 priority patent/US8273724B2/en
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/46449Melanoma antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/31Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/57Skin; melanoma
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies

Definitions

  • This invention is in the field of prevention and treatment of human cancers. More specifically, this invention relates to genes encoding melanoma antigens recognized by T-Cells and their corresponding proteins and to preventative, diagnostic and therapeutic applications which employ these genes or proteins.
  • Melanomas are aggressive, frequently metastatic tumors derived from either melanocytes or melanocyte related nevus cells (“Cellular and Molecular Immunology” (1991) (eds) Abbas A. K., Lechtiman, A. H., Pober, J. S.; W. B. Saunders Company, Philadelphia: pages 340-341). Melanomas make up approximately three percent of all skin cancers and the worldwide increase in melanoma is unsurpassed by any other neoplasm with the exception of lung cancer in women (“Cellular and Molecular Immunology” (1991) (eds) Abbas, A. K., Lechtiman, A. H., Pober, J. S.; W.B.
  • T cells play an important role in tumor regression in most murine tumor models.
  • Tumor infiltrating lymphocytes that recognize unique cancer antigens can be isolated from many murine tumors.
  • the adoptive transfer of these TIL plus interleukin-2 can mediate the regression of established lung and liver metastases (Rosenberg, S. A., et al., (1986) Science 233:1318-1321).
  • the secretion of IFN- ⁇ by injected TIL significantly correlates with in vivo regression of murine tumors suggesting activation of T-cells by the tumor antigens.
  • T cell receptors on CD8 30 T cells recognize a complex consisting of an antigenic peptide (9-10 amino acids for HLA-A2), ⁇ -2 microglobulin and class I major histocompatibility complex (MHC) heavy chain (HLA-A, B, C, in humans). Peptides generated by digestion of endogenously synthesized proteins are transported into the endoplastic reticulum, bound to class I MHC heavy chain and ⁇ 2 microglobulin, and finally expressed in the cell surface in the groove of the class I MHC molecule. Therefore, T cells can detect molecules that originate from proteins inside cells, in contrast to antibodies that detect intact molecules expressed on the cell surface. Therefore, antigens recognized by T cells may be more useful than antigens recognized by antibodies.
  • lymphocytes when isolated, are capable of recognizing specific tumor antigens on autologous and allogeneic melanomas in an MHC restricted fashion.
  • TIL from patients with metastatic melanoma recognize shared antigens including melanocyte-melanoma lineage specific tissue antigens in vitro (Kawakami, Y., et al., (1993) J. Immunother. 14: 88-93; Anichini, A.
  • Anti-melanoma T cells appear to be enriched in TIL probably as a consequence of clonal expansion and accumulation at the tumor site in vivo (Sensi, M., et al., (1993) J. Exp. Med. 178:1231-1246).
  • TAA tumor associated antigens
  • Peripheral blood lymphocytes have been used to identify potential melanoma tumor antigens.
  • Van Der Bruggen et al. (1991) Science 254: 1643-1647 has characterized a gene coding for a melanoma antigen, designated MAGE-1, using T cell clones established from the peripheral blood of patients who were repetitively immunized in vivo with mutagenized tumor cells.
  • Cytotoxic T-cells derived from the peripheral blood lymphocytes of patients with melanoma were used to identify a potential antigenic peptide encoding MAGE-1 (Traversari, C., et al. (1992) J. Exp. Med. 176:1453-1457). Brichard et al. (1993) J. Exp. Med.
  • 178:489-495 has also characterized a gene encoding a melanoma antigen designated tyrosinase using peripheral blood lymphocytes from patients who were sensitized by repetitive in vitro stimulation with tumor. Further support for the therapeutic potential of melanoma antigens is provided by Brown et al. (U.S. Pat. No. 5,262,177). Brown et al. (U.S. Pat. No. 5,262,177) relates to a recombinant vaccinia virus-based melanoma vaccine where the melanoma antigen p97 is reported to show a protective effect from tumor cell challenge in a murine model. Characterization of additional melanoma antigens is important for the development of new strategies for cancer immunotherapy, in particular for melanoma.
  • This invention relates, in general, to nucleic acid sequences encoding melanoma antigens recognized by T-lymphocytes (MART-1) and protein and peptides encoded by these sequences. This invention further provides bioassays for these nucleic acid sequences, proteins and peptides. This invention also provides peptides which have been derived from the MART-1 amino acid sequence and modified to enhance their immunogenocity. This invention also provides therapeutic uses for the nucleic acid sequences, proteins, peptides or modified peptides described herein.
  • FIG. 1 shows the nucleotide and predicted amino acid sequence of the cDNA encoding the MART-1 antigen.
  • the hydrophobic region is underlined.
  • FIG. 2 shows titration of MART-1 peptides for recognition by TIL.
  • T2 cells were incubated with varied concentrations of the purified MART-1 peptides, M9-1, M9-2, M9-3, M10-2, M10-3, M10-4 and M10-5, and lysis by TIL clone A42 or TIL line TIL1235 was measured by 4 h- 51 Cr release cytotoxicity assay at an E (EFFECTOR):T (TARGET) ratio of 20:1 for A42 and 40:1 for TIL1235.
  • E E
  • ECTOR E
  • T TARGET
  • Peptide M9-2 sensitized T2 cells at a concentration of 1 ng/ml.
  • the purified peptide M10-4 was recognized by TIL1235, but not by A42.
  • FIG. 3A shows a radionuclide scan of patient 1200 with metastatic melanoma after receiving the adoptive transfer of autologous 111 In labeled TIL1200.
  • the arrow indicates one of the areas of TIL accumulation corresponding to a metastatic lesion in the left thigh.
  • FIG. 3B shows regression of subcutaneous metastatic tumors following treatment with TIL1200 plus IL-2. Treatment began on day 0.
  • FIG. 4 show the nucleic acid sequence of the full length cDNA25. The start and stop codons are underlined.
  • FIG. 5A shows amino acid sequence of the full length cDNA25. The antigenic peptide is underlined.
  • FIG. 5B shows comparison of the amino acid sequence of the full length cDNA25 (cDNA25FL), the truncated form of cDNA25 (cDNA25TR), Pmel17, ME20 and gp100. (•indicates deletion; - indicates identity).
  • FIG. 6 shows northern blot analysis of melanoma and neonatal melanocyte cell lines and various fresh tissues (10-20 ⁇ g of total RNA) with a cDNA25 probe(the Sal I digested fragment of pCRII-cDNA25) and the ⁇ -actin probe(Clontech).
  • C32, 586mel melanoma cell lines and NHEM529, NHEM530 neonatal melanocyte cell lines were very weak positive.
  • FIGS. 7A-7B show the location of gp100 epitopes and the DNA fragments tested for epitope analysis and recognition by CTL.
  • FIG. 7A Five DNA fragments (D3, D5, D4, C4, 25TR) tested for epitope analysis are shown ( - - - , identical amino acid). Locations of the identified epitopes are underlined.
  • FIG. 7B Recognition by CTL (620-1, 620-2, 660-1, 1143, 1200) of COS 7 cells transfected with each DNA fragment in pcDNA3 plasmid along with HLA-A2.1 cDNA by IFN- ⁇ secretion assays are shown (+, recognized; ⁇ , not recognized).
  • FIGS. 8A-8D show titration of gp100 epitopes by sensitization of HLA-A2.1+T2 cells for CTL lysis. Lysis of T2 cells preincubated with peptides was tested in a 4 h 51 Cr release cytotoxicity assay.
  • FIG. 8A Lysis by TIL1200 of T2 cells incubated with G9 154 ( ⁇ ) or G 10 154 ( ⁇ ).
  • FIG. 8B Lysis by TIL620 of T2 cells incubated with G9 209 ( ⁇ ) or G10 208 ( ⁇ )
  • FIG. 8C Lysis by TIL660-1 of T2 cells incubated with G9 280 ( ⁇ )
  • FIG. 8D Lysis by TIL660-2 of T2 cells incubated with G10-5( ⁇ ).
  • Nucleic acid sequences includes, but is not limited to, DNA, RNA or cDNA.
  • Nucleic acid sequence as used herein refers to an isolated and purified nucleic acid sequence.
  • MART-1 messenger RNA (mRNA) refers to one or more RNA transcripts which are a product of the MART-1 gene.
  • Substantially homologous as used herein refers to substantial correspondence between the nucleic acid sequence of MART-1 shown in FIG. 1 (SEQ ID NO:1) and that of any other nucleic acid sequence.
  • Substantially homologous means about 50-100% homologous homology, preferably by about 70-100% homology, and most preferably about 90-100% homology between the MART-1 sequence and that of any other nucleic acid sequence.
  • substantially homologous as used herein also refers to substantial correspondences between the amino acid sequence of the MART-1 antigen shown in FIG. 1 (SEQ ID NO:2) and that of any other amino acid sequence.
  • MHC Major Histocompatibility Complex
  • HLA human leucocyte antigens
  • melanoma includes, but is not limited to, melanomas, metastatic melanomas, melanomas derived from either melanocytes or melanocytes related nevus cells, melanocarcinomas, melanoepitheliomas, melanosarcomas, melanoma in situ, superficial spreading melanoma, nodular melanoma, lentigo maligna melanoma, acral lentiginous melanoma, invasive melanoma or familial atypical mole and melanoma (FAM-M) syndrome.
  • Such melanomas in mammals may be caused by, chromosomal abnormalities, degenerative growth and developmental disorders, mitogenic agents, ultraviolet radiation (UV), viral infections, inappropriate tissue expression of a gene, alterations in expression of a gene, or carcinogenic agents.
  • UV ultraviolet radiation
  • the aforementioned melanomas can be diagnosed, assessed or treated by methods described in the present application.
  • atypical mole we mean a mole with features that are abnormal and may be precancerous.
  • melanoma antigen or immunogen we mean all or parts thereof of the MART-1 protein or peptides based on the MART-1 protein sequence capable of causing a cellular or humoral immune response in a mammal.
  • antigens may also be reactive with antibodies from animals immunized with all, part or parts of the MART-1 protein (SEQ ID NO:2).
  • SEQ ID NO:2 Such a protein or peptide may be encoded by all or part of the MART-1 nucleic acid sequence of this invention.
  • immunogenic peptide we mean a peptide derived from the MART-1 protein sequence ( FIG. 1 ; SEQ ID NO. 2) or a gp100 protein sequence ( FIG. 5A ; SEQ ID NO: 27) capable of causing a cellular or humoral immune response in a mammal.
  • Such peptides may be reactive with antibodies from an animal immunized with the peptides.
  • Such peptides are about 5-20 amino acid in length preferably about 8 to 15 amino acids in length, and most preferably about 9-10 amino acids in length.
  • bioassays of the present invention may be used in the analysis of biological samples or tissues from any vertebrate species.
  • mammalian biological samples or tissues are analyzed.
  • Tissue includes, but is not limited to, single cells, whole organs and portions thereof.
  • Biological samples include, but are not limited to, tissues, primary cultures of mammalian tissues, biopsy specimens, pathology specimens, and necropsy specimens.
  • Mammal includes but is not limited to, humans, monkeys, dogs, cats, mice, rats, pigs, cows, pigs, horses, sheep and goats.
  • the present invention provides a nucleic acid sequence which encodes a novel melanoma antigen recognized by T cells.
  • This novel melanoma antigen designated MART-1 (melanoma antigen-recognized by T-Cells-1).
  • MART-1 shows no significant homology to any known melanoma antigen and thus represents a new melanoma antigen.
  • the MART-1 antigen contains a highly hydrophobic region from amino acids 27 to 47 (SEQ ID. NO:2) followed by three arginine residues, suggestive of a transmembrane protein. Although no significant homology exists to the entire protein there is a 27 amino acid segment (amino acids 57-83; SEQ ID.
  • MART-1 does not contain a leader sequence characteristic of many Type I membrane proteins (Singer, S. J. (1990) Annu. Rev. Cell Biol. 6: 247-296).
  • MART-1 RNA expression appears to be restricted to fresh and cultured melanoma and melanocyte cell lines and human retina; expression has not been found in any other fresh or cultured tissues or other tumor histologies tested.
  • the cDNA sequence for MART-1 is shown in FIG. 1 (SEQ ID NO:1), the deduced amino acid sequence for the MART-1 protein is also shown in FIG. 1 (SEQ ID NO.:1).
  • the nucleic acid sequence for MART-1 shown in FIG. 1 represents a preferred embodiment of the invention. It is, however, understood by one skilled in the art that due to the degeneracy of the genetic code variations in the cDNA sequence shown in FIG. 1 (SEQ ID NO.:1) will still result in a DNA sequence capable of encoding the MART-1 protein antigen. Such DNA sequences are therefore functionally equivalent to the sequence set forth in FIG. 1 (SEQ ID NO.:1) and are intended to be encompassed within the present invention. Further, a person of skill in the art will understand that there are naturally occurring allelic variations in a given species of the MART-1 nucleic acid sequence shown in FIG. 1 (SEQ ID NO.:1), these variations are also intended to be encompassed by the present invention.
  • the predicted MART-1 antigen is a 118 amino acid protein of about 13 (kd).
  • This invention further includes MART-1 protein or peptides or analogs thereof having substantially the same function as the MART-1 antigen or protein of this invention.
  • Such proteins or polypeptides include, but are not limited to, a fragment of the protein, or a substitution, addition or deletion mutant of the MART-1 protein.
  • This invention also encompasses proteins or peptides that are substantially homologous to the MART-1 antigen. Substantially homologous means about 50-100% homology, preferably about 70-100% homology, and most preferably about 90-100% homology between the MART-1 and any another amino acid sequence or protein or peptide.
  • analog includes any polypeptide having an amino acid residue sequence substantially identical to the MART-1 sequence specifically shown herein ( FIG. 1 ; SEQ ID NO: 1) in which one or more residues have been conservatively substituted with a functionally similar residue and which displays the functional aspects of the MART-1 antigen as described herein.
  • conservative substitutions include the substitution of one non-polar (hydrophobic) residue such as isoleucine, valine, leucine or methionine for another, the substitution of one polar (hydrophilic) residue for another such as between arginine and lysine, between glutamine and asparagine, between glycine and serine, the substitution of one basic residue such as lysine, arginine or histidine for another, or the substitution of one acidic residue, such as aspartic acid or glutamic acid or another.
  • “conservative substitution” also includes the use of a chemically derivatized residue in place of a non-derivatized residue.
  • “Chemical derivative” refers to a subject polypeptide having one or more residues chemically derivatized by reaction of a functional side group. Examples of such derivatized molecules include for example, those molecules in which free amino groups have been derivatized to form amine hydrochlorides, p-toluene sulfonyl groups, carbobenzoxy groups, t-butyloxycarbonyl groups, chloroacetyl groups or formyl groups. Free carboxyl groups may be derivatized to form salts, methyl and ethyl esters or other types of esters or hydrazides.
  • Free hydroxyl groups may be derivatized to form O-acyl or O-alkyl derivatives.
  • the imidazole nitrogen of histidine may be derivatized to form N-im-benzylhistidine.
  • chemical derivatives are those proteins or peptides which contain one or more naturally-occurring amino acid derivatives of the twenty standard amino acids. For examples: 4-hydroxyproline may be substituted for proline; 5-hydroxylysine may be substituted for lysine; 3-methylhistidine may be substituted for histidine; homoserine may be substituted for serine; and ornithine may be substituted for lysine.
  • Proteins or polypeptides of the present invention also include any polypeptide having one or more additions and/or deletions or residues relative to the sequence of a polypeptide whose sequence is encoded is the DNA of MART-1, so long as the requisite activity is maintained.
  • This invention also provides a recombinant DNA molecule comprising all or part of the MART-1 nucleic acid sequence (SEQ. ID NO.:1) and a vector.
  • Expression vectors suitable for use in the present invention may comprise at least one expression control element operationally linked to the nucleic acid sequence.
  • the expression control elements are inserted in the vector to control and regulate the expression of the nucleic acid sequence. Examples of expression control elements include, but are not limited to, lac system, operator and promoter regions of phage lambda, yeast promoters and promoters derived from polyoma, adenovirus, retrovirus or SV40.
  • Additional preferred or required operational elements include, but are not limited to, leader sequence, termination codons, polyadenylation signals and any other sequences necessary or preferred for the appropriate transcription and subsequent translation of the nucleic acid sequence in the host system. It will be understood by one skilled in the art the correct combination of required or preferred expression control elements will depend on the host system chosen. It will further be understood that the expression vector should contain additional elements necessary for the transfer and subsequent replication of the expression vector containing the nucleic acid sequence in the host system. Examples of such elements include, but are not limited to, origins of replication and selectable markers.
  • Another aspect of this invention relates to a host organism into which recombinant expression vector containing all or part of the MART-1 nucleic acid sequence has been inserted.
  • the host cells transformed with the MART-1 nucleic acid sequence of this invention include eukaryotes, such as animal, plant, insect and yeast cells and prokaryotes, such as E. coli.
  • the means by which the vector carrying the gene may be introduced into the cell include, but are not limited to, microinjection, electroporation, transduction, or transfection using DEAE-dextran, lipofection, calcium phosphate or other procedures known to one skilled in the art (Sambrook et al. (1989) in “Molecular Cloning. A Laboratory Manual”, Cold Spring Harbor Press, Plainview, N.Y.).
  • eukaryotic expression vectors that function in eukaryotic cells are used.
  • examples of such vectors include, but are not limited to, retroviral vectors, vaccinia virus vectors, adenovirus vectors, herpes virus vector, fowl pox virus vector, bacterial expression vectors, plasmids, such as pcDNA3 (Invitrogen, San Diego, Calif.) or the baculovirus transfer vectors.
  • Preferred eukaryotic cell lines include, but are not limited to, COS cells, CHO cells, HeLa cells, NIH/3T3 cells, 293 cells (ATCC#CRL1573), T2 cells, dendritic cells, or monocytes.
  • the recombinant MART-1 protein expression vector is introduced into mammalian cells, such as NIH/3T3, COS, CHO, 293 cells (ATCC #CRL 1573), T2 cells, dendritic cells, or monocytes to ensure proper processing and modification of the MART-1 protein.
  • mammalian cells such as NIH/3T3, COS, CHO, 293 cells (ATCC #CRL 1573), T2 cells, dendritic cells, or monocytes.
  • the MART-1 cDNA is introduced into COS7 (Gluzman, Y. et al. (1981) Cell 23: 175-182). The choice of an appropriate cell is within the skill of a person in the art.
  • the expressed recombinant MART-1 protein may be detected by methods known in the art which include Coomassie blue staining and Western blotting using antibodies specific for the MART-1 protein.
  • the recombinant protein expressed by the host cells can be obtained as a crude lysate or can be purified by standard protein purification procedures known in the art which may include differential precipitation, molecular sieve chromatography, ion-exchange chromatography, isoelectric focusing, gel electrophoresis, affinity, and immunoaffinity chromatography and the like.
  • standard protein purification procedures known in the art which may include differential precipitation, molecular sieve chromatography, ion-exchange chromatography, isoelectric focusing, gel electrophoresis, affinity, and immunoaffinity chromatography and the like.
  • immunoaffinity chromatography the recombinant protein may be purified by passage through a column containing a resin which has bound thereto antibodies specific for the MART-1 protein (Ausubel et. al., (1987) in “Current Protocols in Molecular Biology” John Wiley and Sons, New York, N.Y.).
  • nucleic acid sequence or portions thereof, of this invention are useful as probes for the detection of expression of the MART-1 gene in normal and diseased tissue. Therefore, another aspect of the present invention relates to a bioassay for detecting messenger RNA encoding the MART-1 protein in a biological sample comprising the steps of (a) contacting all or part of the nucleic acid sequence of this invention with said biological sample under conditions allowing a complex to form between said nucleic acid sequence and said messenger RNA, (b) detecting said complexes and, (c) determining the level of said messenger RNA.
  • RNA can be isolated as whole cell RNA or as poly(A) + RNA.
  • Whole cell RNA can be isolated by a variety of methods known to those skilled in the art. (Ausubel et al., (1987) on “Current Protocols in Molecular Biology”, John Wiley and Sons, New York). Such methods include extraction of RNA by differential precipitation (Birnboim, H. C. (1988) Nucleic Acids Res., 16:1487-1497), extraction of RNA by organic solvents (Chomczynski, P. et al. (1987) Anal. Biochem., 162:156-159) and the extraction of RNA with strong denaturants (Chirgwin, J. M. et al.
  • Poly(A) + RNA can be selected from whole cell RNA by affinity chromatography on oligo-d(T) columns (Aviv, H. et al. (1972) Proc. Natl. Acad. Sci., 69:1408-1412).
  • Examples of methods for determining cellular messenger mRNA levels for step (c) include, but are not limited to Northern blotting (Alwine, J. C. et al. (1977) Proc. Natl. Acad. Sci., 74:5350-5354), dot and slot hybridization (Kafatos, F. C. et al.
  • Detection of complexes in Step (b) of the bioassay can also be carried out by a variety of techniques. Detection of the complexes by signal amplification can be achieved by several conventional labelling techniques including radiolabels and enzymes (Sambrook et. al., (1983) in “Molecular Cloning, A Laboratory Manual”, Cold Spring Harbor Press, Plainview, N.Y.; Ausubel et al., (1987) in “Current Protocols in Molecular Biology, John Wiley and Sons, New York N.Y.). Radiolabelling kits are also commercially available.
  • the MART-1 nucleic acid sequence used as a probe in step(c) of the bioassay may be RNA or DNA.
  • Preferred methods of labelling the DNA sequences are with 32 P using Klenow enzyme or polynucleotide kinase.
  • Preferred methods of labeling RNA or riboprobe sequences are with 32 P or 35 S using RNA polymerases.
  • non-radioactive techniques for signal amplification including methods for attaching chemical moieties to pyrimidine and purine rings (Dale, R. N. K. et al. (1973) Proc. Natl. Acad. Sci., 70:2238-2242; Heck, R. F. (1968) S. Am. Chem. Soc., 90:5518-5523), methods which allow detection by chemiluminescence (Barton, S. K. et al.
  • biological samples that can be used in this bioassay include, but are not limited to, primary mammalian cultures, continuous mammalian cell lines, such as melanocyte cell lines, mammalian organs such as skin or retina, tissues, biopsy specimens, neoplasms, pathology specimens, and necropsy specimens.
  • a 32 P radiolabelled MART-1 probe is used.
  • the MART-1 probe is the full length cDNA in FIG. 1 (SEQ ID NO:1).
  • the approximately 1.6 Kilobase (kb) cDNA ( FIG. 1 ; SEQ ID NO:1) was cloned into the vector and the resulting plasmid deposited with the American Type Culture Collection (ATCC) 12301 Parklawn Drive, Rockville, Md. 20852 USA on Apr. 14, 1994, and given ATCC Deposit Number 75738.
  • the full length MART-1 nucleic acid sequence can be isolated from the pCRII plasmid by digestion with HINDIII and XhoI restriction enzymes. This 1.6 kb nucleic acid sequence can then be used as a probe.
  • This probe is used to detect MART-1 mRNA in total RNA or poly A + RNA isolated from a variety of tissues or biological samples.
  • oligonucleotide pairs based on the MART-1 sequence in FIG. 1 are used as Polymerase Chain Reaction (PCR) primers to detect MART-1 mRNA in a biological sample.
  • PCR Polymerase Chain Reaction
  • These primers can be used in a method following the reverse transcriptase-Polymerase Chain Reaction (RT-PCR) process for amplifying selected RNA nucleic acid sequences as detailed in Ausubel et al., (eds) (1987) In “Current Protocols in Molecular Biology” Chapter 15, John Wiley and Sons, New York, N.Y.
  • oligonucleotides can be synthesized by automated instruments sold by a variety of manufacturers or can be commercially prepared based upon the nucleic acid sequence of this invention.
  • One skilled in the art will know how to select PCR primers based on the MART-1 nucleic acid sequence for amplifying MART-1 RNA in a sample.
  • the MART-1 nucleic acid sequence or portions thereof are useful to detect alterations of the MART-1 gene in normal or diseased mammalian tissue.
  • alteration we mean additions, deletions, substitutions or duplications in the MART-1 gene sequence or gene amplification of the MART-1 gene sequence.
  • another aspect of the present invention relates to an assay for detecting alterations of the MART-1 gene in a biological sample comprising the steps of (a) contacting all or part of the nucleic acid sequence of this invention with genomic DNA isolated from a biological sample under conditions allowing a complex to form between said nucleic acid sequence and said genomic DNA, (b) detecting said complexes, and (c) determining alterations in said MART-1 gene by comparison to a control sample.
  • the MART-1 nucleic acid sequences of this invention can also be used as probes to isolate the MART-1 homologs in other species.
  • the MART-1 cDNA ( FIG. 1 ; SEQ ID No:1) is used to screen a mammalian cDNA library, positive clones are selected and sequenced.
  • tissue sources from which the cDNA library can be synthesized include, but are not limited to skin, retina, melanocytes, neonatal skin and embryos.
  • a melanoma library is screened using the MART-1 cDNA as a probe ( FIG. 1 ; SEQ ID No. 1).
  • MART-1 protein is an antigen present on melanoma cells. It is therefore another aspect of this invention to provide MART-1 nucleic acid probes to be utilized in detecting MART-1 RNA or alterations in the level of MART-1 mRNA in biological sample isolated from a mammal afflicted with a disease. Examples of such diseases, include but are not limited to, melanomas.
  • alterations in the level of MART-1 mRNA we mean an increase or decrease in the level of an RNA relative to a control sample or the appearance or disappearance of the MART-1 mRNA relative to a control sample. Detection in the alterations of MART-1 mRNA will allow for diagnosis or the assessment of the diseased state. Therefore, alterations in the level of MART-1 mRNA may be predictive of the prognosis for the afflicted mammal.
  • the nucleic acid of this invention can be used in in situ hybridization on mammalian tissues to determine the precise site or subcellular site of expression of the MART-1 gene within a tissue.
  • a preferred method of labeling the MART-1 nucleic acid sequence is synthesizing a 35 S-labeled RNA probe by in vitro transcription utilizing SP6 polymerase.
  • the sense strand is under the control of the T7 promoter
  • the antisense strand is under the SP6 promoter. It is preferable that the probe be hydrolyzed to a probe length of approximately 400-200 base pairs.
  • tissues examples include, but are not limited to, mammalian embryos, adult mammalian tissues, such as skin, lymph nodes and retina, biopsy specimens, pathology specimens and necropsy specimens.
  • MART-1 in situ probes may be used to evaluate MART-1 RNA expression in diseased tissue for invasive early melanoma to characterize radial and vertical growth phases of the melanoma lesion and assess the margins of the disease within the tissue.
  • all or parts thereof of the MART-1 (SEQ ID NO:1) nucleic acid sequence can be used to generate transgenic animals.
  • the MART-1 gene is introduced into an animal or an ancestor of the animal at an embryonic stage, preferably at the one cell stage and generally not later than about the eight cell stage.
  • transgenic animals carrying a MART-1 gene can be made.
  • One method involves the use of retroviruses carrying all or part of the MART-1 sequence.
  • the retroviruses containing the transgene are introduced into the embryonic animal by transfection.
  • Another methods involves directly injecting the transgene into the embryo.
  • Yet another methods employs the embryonic stem cell method or homologous recombination method known to workers in the field.
  • Examples of animals into which the MART-1 transgene can be introduced include but are not limited to, primates, mice, rats or other rodents. Such transgenic animals may be useful as biological models for the study of melanoma and to evaluate diagnostic or therapeutic methods for melanoma.
  • This invention further comprises an antibody or antibodies reactive with the MART-1 protein or peptides or modified peptides or analogs thereof having the amino acid sequence defined in FIG. 1 (SEQ ID NO: 2) or a unique portion thereof.
  • the antibodies are monoclonal or polyclonal in origin.
  • MART-1 protein or peptides used to generate the antibodies may be from natural or recombinant sources or generated by chemical synthesis.
  • Natural MART-1 proteins can be isolated from mammalian biological samples. Biological samples include, but are not limited to mammalian tissues such as fresh melanoma, skin, retina, primary or continuous cultures of mammalian cells such as melanoma cultures or cultured melanocytes.
  • the natural MART-1 proteins may be isolated by the same methods described above for recombinant proteins.
  • Recombinant MART-1 proteins or peptides may be produced and purified by conventional methods.
  • Synthetic MART-1 peptides may be custom ordered or commercially made based on the predicted amino acid sequence of the present invention ( FIG. 1 ; SEQ ID:2) or synthesized by methods known to one skilled in the art (Merrifield, R. B. (1963) J. Amer. Soc. 85:2149).
  • Examples of MART-1 peptides include, but are not limited to, are AAGIGILTV (M9-2; SEQ. ID NO. 4), EAAGIGILTV (M10-3; SEQ. ID NO. 17) and AAGIGILTVI (M10-4; SEQ. ID NO. 18) (peptides are presented in single letter amino acid code).
  • the most preferred peptide is AAGIGILTV (SEQ ID NO:4).
  • peptides derived form the MART-1 protein sequence may modified to increase their immunogenicity by enhancing binding of the peptide to the MHC molecules in which the peptide is presented.
  • modified MART-1 peptides that may used are shown, but not limited to, the peptides in Table 14.
  • the MART-1 peptide that is modified to enhance its binding to MHC Class I molecules is AAGIGILTV (SEQ ID NO:4).
  • the peptide or modified peptide may be conjugated to a carrier molecule to enhance the antigenicity of the peptide.
  • carrier molecules include, but are not limited to, human albumin, bovine albumin, lipoprotein and keyhole limpet hemo-cyanin (“Basic and Clinical Immunology” (1991) Stites, D. P. and Terr A. I. (eds) Appleton and Lange, Norwalk Connecticut, San Mateo, Calif.).
  • Exemplary antibody molecules for use in the detection methods of the present invention are intact immunoglobulin molecules, substantially intact immunoglobulin molecules or those portions of an immunoglobulin molecule that contain the antigen binding site, including those portions of immunoglobulin molecules known in the art as F(ab), F(ab′); F(ab) 2 and F(v).
  • Polyclonal or monoclonal antibodies may be produced by methods known in the art. (Kohler and Milstein (1975) Nature 256, 495-497; Campbell “Monoclonal Antibody Technology, the Production and Characterization of Rodent and Human Hybridomas” in Burdon et al.
  • the antibodies of this invention may react with native or denatured MART-1 protein, peptides or analogs thereof, or modified peptides an analogs thereof.
  • the specific immunoassay in which the antibodies are to be used will dictate which antibodies are desirable.
  • Antibodies may be raised against the MART-1 protein or portions thereof or against synthetic peptides homologous to the MART-1 amino acid sequence.
  • the antibodies of this invention are used in immunoassays to detect the novel MART-1 protein in biological samples.
  • the antibodies of the present invention are contacted with a biological sample and the formation of a complex between the MART-1 antigen and antibody is detected.
  • Immunoassays of the present invention may be radioimmunoassay, Western blot assay, immunofluorescent assay, enzyme immunoassay, chemiluminescent assay, immunohistochemical assay and the like.
  • Neoman (eds), Stockton Pres, NY, N.Y.; Oellirich, M. 1984. J. Clin. Chem. Clin. Biochem. 22: 895-904)
  • Biological samples appropriate for such detection assays include mammalian tissues, melanoma and melanocyte cell lines, skin, retina, lymph nodes, pathology specimens, necropsy specimens, and biopsy specimens. Proteins may be isolated from biological samples by conventional methods described in (Ausubel et al., (eds) (1987) in “Current Protocols in Molecular Biology” John Wiley and Sons, New York, N.Y.).
  • the antibodies of this invention can therefore be used in immunoassays to detect MART-1 antigen or alteration in the level of expression of the MART-1 antigen in biological samples isolated from mammals afflicted with a disease or disorder.
  • biological samples include, but are not limited to, mammalian tissues, biopsy tissue samples, melanoma and lymph node biopsy samples, pathology and tissue samples.
  • diseases that can be assessed by these immunoassays, include, but are not limited to, melanomas and tissues which are secondary sites for melanoma metastasis.
  • alteration in level of expression we mean an increase or decrease of the MART protein or portions thereof relative to a control sample. Alteration is also meant to encompass substitution, deletion or addition mutants of the MART-1 protein.
  • Such mutations can be determined by using the antibodies of this invention known to react with specific epitopes of the MART-1 protein and determining which epitopes are present relative to a control.
  • the antibodies of this invention can therefore be used in an immunoassay to diagnose, assess or prognoses a mammal afflicted with the disease.
  • the MART-1 antibodies of this invention are used to assess the presence of the MART-1 antigen from a tissue biopsy of a mammal afflicted with melanoma using immunocytochemistry. Such assessment of the delineation of the MART-1 antigen in a diseased tissue can be used to prognose the progression of the disease in a mammal afflicted with the disease.
  • the MART-1 antibodies can be used to characterize the radial and vertical growth phases of the melanoma lesion. Conventional methods for immunohistochemistry are described in (Harlow and Lane (eds) (1988) In “Antibodies A Laboratory Manual”, Cold Spinning Harbor Press, Cold Spring Harbor, N.Y.; Ausbel et al. (eds) (1987). In Current Protocols In Molecular Biology John Wiley and Sons (New York, N.Y.).
  • antibodies of this invention may be used to purify the MART-1 protein or portions thereof.
  • Immunoaffinity chromatography can be performed by conventional methods known to one skilled in the art (Ausubel et al. (eds) (1987) in “Current Protocols in Molecular Biology” John Wiley and Sons, New York, N.Y.).
  • rabbit antisera containing antibodies which specifically recognize the MART-1 protein is used to detect said protein in Western Blot Analysis.
  • Such antisera is directed to all, or a part or parts of the MART-1 protein or synthetic peptides derived from the MART-1 protein sequence.
  • a MART-1 synthetic peptide derived from the MART-1 predicted amino acid sequence is used ( FIG. 1 ; SEQ ID NO:2).
  • modified MART-1 peptides may be used.
  • the peptide is synthesized by standard methods on an automated peptide synthesizer and purified by high pressure liquid chromatography (HPLC) as described in Example 2.
  • HPLC high pressure liquid chromatography
  • the purified peptide may be conjugated to a carrier as described in (M.
  • rabbits may be immunized with the MART-1 protein or peptide conjugated to carriers.
  • the animal receives similar booster doses and antisera titer is assessed by ELISA assay. Satisfactory levels of antisera are obtained when the anti-peptide antibody titer reaches a plateau. This antibody can be used in the standard immunoassays described above.
  • T-lymphocytes recognize antigen in association with Class I or Class II MHC molecules in the form of a peptide fragment bound to an MHC molecule.
  • the degree of peptide binding to a given MHC allele is based on amino acids at particular positions within the peptide (Parker et al. (1992) Journal of Immunology 149:3580; Kubo, et al. (1994) Journal of Immunology 52:3913-3924; Ruppert J. et al. (1993) Cell 74:929-937; Falk et al. (1991) Nature 351:290-296, each of which is herein incorporated by reference). Therefore, another embodiment of this invention relates to peptides derived from the MART-1 protein sequence ( FIG.
  • modification may include substitution, deletion or addition of an amino acid in the given-immunogenic peptide sequence or mutation of existing amino acids within the given immunogenic peptide sequence, or derivatization of existing amino acids within the given immunogenic peptide sequence.
  • Any amino acid comprising the immunogenic peptide sequence may be modified in accordance with this invention. In a preferred embodiment at least one amino acid is substituted or replaced within the given immunogenic peptide sequence. Any amino acid may be used to substitute or replace a given amino acid within the immunogenic peptide sequence.
  • Modified peptides are intended to include any immunogenic MART-1 peptide which has been modified and exhibits enhanced binding to the MHC molecule with which it associates when presented to the T-cell.
  • the HLA-A2 allele binds peptides of nine or ten amino acids.
  • positions within the peptide that may be altered to enhance binding include, but are not limited to, the first position, the second position, the third position and the last position of the peptide. Any amino acid may be used to substitute or replace these positions within the immunogenic peptide sequence.
  • the amino acid at the second position of the peptide is preferably a hydrophobia aliphatic amino acid.
  • Examples or amino acids that may be used at the second position include, but are not limited to, leucine, methionine, alanine, isoleucine, valine, threonine or glycine.
  • leucine or methionine is found at the second position of the peptide.
  • the last amino acid of the peptide is preferably a hydrophobic aliphatic amino acid.
  • amino acids that may be used in the last position of the peptide include, but are not limited to, valine, methionine, leucine, alanine, isoleucine, threonine or glycine.
  • valine is found at the last position in the peptide.
  • the amino acids at the first and third positions in the peptide may also be modified to enhance binding of the peptide to the MHC Class I molecule.
  • the amino acids at the first and third positions in the peptide may be any amino acid.
  • the amino acids at the first and third positions are hydrophobic aliphatic amino acids or aromatic amino acids.
  • Examples of amino acids that maybe used at these positions include, but are not limited to, leucine, methionine, valine, alanine, isoleucine, threonine, glycine, tryptophan, phenylalanine, tyrosine, serine, aspartic acid, or lysine.
  • MART-1 peptides examples include, but are not limited to, AAGIGILTV (SEQ ID NO: 4), EAAGIGILTV (SEQ ID NO: 17) and AAGIGILTVI (SEQ ID NO: 18) (peptides are presented in single letter amino acid code).
  • AAGIGILTV SEQ ID NO: 4
  • EAAGIGILTV SEQ ID NO: 17
  • AAGIGILTVI SEQ ID NO: 18
  • the immunogenic MART-1 peptide AAGIGILTV may be modified according to the following formula X 1 X 2 X 3 IGILTX 4 wherein
  • X 1 may be any amino acid, preferably any hydrophobic aliphatic amino acid, or aromatic amino acid.
  • amino acids that may be used, but are not limited to, alanine, tryptophan, phenylalanine, tyrosine, lysine, isoleucine, leucine, methionine, threonine, glycine or serine.
  • X 2 may be any hydrophobic amino acid, preferably an aliphatic hydrophobic amino acids.
  • amino acids that may be used include, but are not limited to, leucine, methionine, isoleucine, valine, threonine, alanine or glycine.
  • X 3 may be any amino acid, preferably any hydrophobic aliphatic amino acid, or aromatic amino acid.
  • amino acids that may be used include, but are not limited to, tryptophan, phenylalanine, tyrosine, lysine, aspartic acid, serine, alanine, glycine, isoleucine, valine, or threonine.
  • X 4 may be any hydropbic amino acid, preferably a hydrophobic aliphatic amino acid.
  • amino acids that may be used include, but are not limited to, valine, leucine, isoleucine, alanine, threonine, or glycine.
  • modified AAGIGILTV SEQ ID NO:4
  • SEQ ID NO:4 Examples of modified AAGIGILTV (SEQ ID NO:4) peptide sequences that may be produced are shown but not limited to the peptides in Table 14 (Example 5).
  • This invention further includes analogs of these immunogenic modified peptides derived from the MART-1 amino acid sequence ( FIG. 1 ; SEQ ID NO:2) which have been modified.
  • the term analog is intended to include any peptide which displays the functional aspects of these modified peptides.
  • the term analog also includes conservative substitutions or chemical derivatives of these modified peptides as described above. These modified peptides may be synthetically or recombinantly produced by conventional methods.
  • the recombinant or natural MART-1 protein, peptides, or analogs thereof, or modified MART-1 peptides, or analogs thereof may be used as a vaccine either prophylactically or therapeutically.
  • the vaccine is provided in advance of any evidence of melanoma.
  • the prophylactic administration of the MART-1 vaccine should serve to prevent or attenuate melanoma in a mammal.
  • mammals, preferably human, at high risk for melanoma are prophylactically treated with the vaccines of this invention.
  • the vaccine is provided to enhance the patient's own immune response to the tumor antigen present on the melanoma or metastatic melanoma.
  • the vaccine which acts as an immunogen, may be a cell, cell lysate from cells transfected with a recombinant expression vector, cell lysates from cells transfected with a MART-1 recombinant expression vector, or a culture supernatant containing the expressed protein.
  • the immunogen is a partially or substantially purified recombinant MART-1 protein, peptide or analog thereof or modified peptides or analogs thereof.
  • the proteins or peptides may be conjugated with lipoprotein or administered in liposomal form or with adjuvant.
  • the immunogen While it is possible for the immunogen to be administered in a pure or substantially pure form, it is preferable to present it as a pharmaceutical composition, formulation or preparation.
  • formulations of the present invention both for veterinary and for human use, comprise an immunogen as described above, together with one or more pharmaceutically acceptable carriers and, optionally, other therapeutic ingredients.
  • the carrier(s) must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any method well-known in the pharmaceutical art.
  • All methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients.
  • the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product into the desired formulation.
  • Formulations suitable for intravenous intramuscular, subcutaneous, or intraperitoneal administration conveniently comprise sterile aqueous solutions of the active ingredient with solutions which are preferably isotonic with the blood of the recipient.
  • Such formulations may be conveniently prepared by dissolving solid active ingredient in water containing physiologically compatible substances such as sodium chloride (e.g. 0.1-2.0M), glycine, and the like, and having a buffered pH compatible with physiological conditions to produce an aqueous solution, and rendering said solution sterile.
  • physiologically compatible substances such as sodium chloride (e.g. 0.1-2.0M), glycine, and the like
  • physiologically compatible substances such as sodium chloride (e.g. 0.1-2.0M), glycine, and the like
  • pH compatible substances such as sodium chloride (e.g. 0.1-2.0M), glycine, and the like
  • These may be present in unit or multi-dose containers, for example, sealed ampoules or vials.
  • the formulations of the present invention may incorporate a stabilizer.
  • Illustrative stabilizers are polyethylene glycol, proteins, saccharides, amino acids, inorganic acids, and organic acids which may be used either on their own or as admixtures. These stabilizers are preferably incorporated in an amount of 0.11-10,000 parts by weight per part by weight of immunogen. If two or more stabilizers are to be used, their total amount is preferably within the range specified above. These stabilizers are used in aqueous solutions at the appropriate concentration and pH. The specific osmotic pressure of such aqueous solutions is generally in the range of 0.1-3.0 osmoles, preferably in the range of 0.8-1.2. The pH of the aqueous solution is adjusted to be within the range of 5.0-9.0, preferably within the range of 6-8. In formulating the immunogen of the present invention, anti-adsorption agent may be used.
  • Controlled release preparations may be achieved through the use of polymer to complex or absorb the proteins or their derivatives.
  • the controlled delivery may be exercised by selecting appropriate macromolecules (for example polyester, polyamino acids, polyvinyl, pyrrolidone, ethylenevinylacetate, methylcellulose, carboxymethylcellulose, or protamine sulfate) and the concentration of macromolecules as well as the methods of incorporation in order to control release.
  • Another possible method to control the duration of action by controlled-release preparations is to incorporate the MART-1 protein, peptides and analogs thereof into particles of a polymeric material such as polyesters, polyamino acids, hydrogels, poly(lactic acid) or ethylene vinylacetate copolymers.
  • microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly(methylmethacylate) microcapsules, respectively, or in colloidal drug delivery systems, for example, liposomes, albumin microspheres, microemulsions, nanoparticles, and nanocapsules or in macroemulsions.
  • compositions may be combined with typical carriers, such as lactose, sucrose, starch, talc magnesium stearate, crystalline cellulose, methyl cellulose, carboxymethyl cellulose, glycerin, sodium alginate or gum arabic among others.
  • typical carriers such as lactose, sucrose, starch, talc magnesium stearate, crystalline cellulose, methyl cellulose, carboxymethyl cellulose, glycerin, sodium alginate or gum arabic among others.
  • the proteins of the present invention may be supplied in the form of a kit, alone, or in the form of a pharmaceutical composition as described above.
  • Vaccination can be conducted by conventional methods.
  • the immunogen can be used in a suitable diluent such as saline or water, or complete or incomplete adjuvants.
  • the immunogen may or may not be bound to a carrier to make the protein immunogenic.
  • carrier molecules include but are not limited to bovine serum albumin (BSA), keyhole limpet hemocyanin (KLH), tetanus toxoid, and the like.
  • BSA bovine serum albumin
  • KLH keyhole limpet hemocyanin
  • the immunogen also may be coupled with lipoproteins or administered in liposomal form or with adjuvants.
  • the immunogen can be administered by any route-appropriate for antibody production such as intravenous, intraperitoneal, intramuscular, subcutaneous, and the like.
  • the immunogen may be administered once or at periodic intervals until a significant titer of anti-MART-1 immune cells or anti-MART-1 antibody is produced.
  • the presence of anti-MART-1 immune cells may be assessed by measuring the frequency of precursor CTL (cytoxic T-lymphocytes) against MART-1 antigen prior to and after immunization by a CTL precursor analysis assay (Coulie, P. et al., (1992) International Journal Of Cancer 50:289-297).
  • the antibody may be detected in the serum using the immunoassay described above.
  • the administration of the vaccine or immunogen of the present invention may be for either a prophylactic or therapeutic purpose.
  • the immunogen is provided in advance of any evidence or in advance of any symptom due to melanoma.
  • the prophylactic administration of the immunogen serves to prevent or attenuate melanoma in a mammal.
  • the immunogen is provided at (or shortly after) the onset of the disease or at the onset of any symptom of the disease.
  • the therapeutic administration of the immunogen serves to attenuate the disease.
  • a preferred embodiment is a vaccine prepared using recombinant MART-1 protein or peptide expression vectors.
  • a genetic sequence which encodes for all or part of the MART-1 nucleic acid sequence is inserted into a expression vector, as described above, and introduced into the mammal to be immunized.
  • vectors that may be used in the aforementioned vaccines include, but are not limited to, defective retroviral vectors, adenoviral vectors vaccinia viral vectors, fowl pox viral vectors, or other viral vectors (Mulligan, R. C., (1993) Science 260:926-932).
  • the viral vectors carrying all or part of the MART-1 nucleic sequence can be introduced into a mammal either prior to any evidence of melanoma or to mediate regression of the disease in a mammal afflicted with melanoma.
  • methods for administering the viral vector into the mammals include, but are not limited to, exposure of cells to the virus ex vivo, or injection of the retrovirus or a producer cell line of the virus into the affected tissue or intravenous administration of the virus.
  • the viral vector carrying all or part of the MART-1 nucleic acid sequence may be administered locally by direct injection into the melanoma lesion or topical application in a pharmaceutically acceptable carrier.
  • the quantity of viral vector, carrying all or part of the MART-1 nucleic acid sequence, to be administered is based on the titer of virus particles.
  • a preferred range of the immunogen to be administered may be about 10 6 to about 10 11 virus particles per mammal, preferably a human.
  • the efficacy of the vaccine can be assessed by production of antibodies or immune cells that recognize the antigen, as assessed by specific lytic activity or specific cytokine production or by tumor regression.
  • One skilled in the art would know the conventional methods to assess the aforementioned parameters. If the mammal to be immunized is already afflicted with melanoma or metastatic melanoma the vaccine can be administered in conjunction with other therapeutic treatments. Examples of other therapeutic treatments includes, but are not limited to, adoptive T cell immunotherapy, coadministration of cytokines or other therapeutic drugs for melanoma.
  • the MART-1 protein may be administered as a vaccine in a pharmaceutically acceptable carrier.
  • Ranges of MART-1 protein that may be administered are about 0.001 to about 100 mg per patient, preferred doses are about 0.01 to about 100 mg per patient.
  • the MART-1 peptide AAGIGILTV (SEQ. ID NO. 4) (presented in single letter code) or analogs thereof is administered therapeutically or prophylactically to a mammal in need of such treatment.
  • modified MART-1 peptides examples of which are presented in Table 14 may be used.
  • Preferred doses may be about 0.001 mg to about 100 mg, most preferred are about 0.01 mg to about 100 mg.
  • the peptide may be synthetically or recombinantly produced. Immunization is repeated as necessary, until a sufficient titer of anti-immunogen antibody or immune cells has been obtained.
  • a viral vector such as a retroviral vector
  • mammalian cells into which the retroviral vector can be introduced include, but are not limited to, primary mammalian cultures or continuous mammalian cultures, COS cells, NIH3T3, or 293 cells (ATTC #CRL 1573).
  • the means by which the vector carrying the gene may be introduced into a cell includes, but is not limited to, microinjection, electroporation, transfection or transfection using DEAE dextran, lipofection, calcium phosphate or other procedures known to one skilled in the art (Sambrook et al. (EDS) (1989) in “Molecular Cloning.
  • the mammalian cells expressing the MART-1 antigen can be administered to mammals and serve as a vaccine or immunogen.
  • Examples of how the cells expressing MART-1 antigens can be administered include, but is not limited to, intravenous, intraperitoneal or intralesional.
  • the part of the MART-1 nucleic acid sequence corresponding to the peptide AAGIGILTV (SEQ ID NO: 4) is inserted into the MART-1 expression vector and introduced into the mammalian cells.
  • a nucleic acid sequence corresponding to MART-1 peptides which have been modified to enhance their binding to MHC molecules may be used.
  • the nucleic acid sequences encoding the modified peptides shown in Table 14 may be inserted into an expressions vector and introduced with mammalian cells.
  • the vaccine formulation of the present invention comprise an immunogen that induces an immune response directed against the melanoma associated antigens such as the melanoma associated MART-1 antigen.
  • the vaccine formulations may be evaluated first in animal models, initially rodents, and in nonhuman primates and finally in humans. The safety of the immunization procedures is determined by looking for the effect of immunization on the general health of the immunized animal (weight change, fever, appetite behavior etc.) and looking for pathological changes on autopsies. After initial testing in animals, melanoma cancer patients can be tested. Conventional methods would be used to evaluate the immune response of the patient to determine the efficiency of the vaccine.
  • all, part, or parts of the MART-1 protein or MART-1 peptides or analogs thereof, or modified MART-1 peptides or analogs thereof, may be exposed to dendritic cells cultured in vitro.
  • the cultured dendritic cells provide a means of producing T-cell dependent antigens comprised of dendritic cell modified antigen or dendritic cells pulsed with antigen, in which the antigen is processed and expressed on the antigen activated dendritic cell.
  • the MART-1 antigen activated dendritic cells or processed dendritic cell antigens may be used as immunogens for vaccines or for the treatment of melanoma.
  • the dendritic cells should be exposed to antigen for sufficient time to allow the antigens to be internalized and presented on the dendritic cells surface.
  • the resulting dendritic cells or the dendritic cell process antigens can than be administered to an individual in need of therapy.
  • Such methods are described in Steinman et al. (WO93/208185) and in Banchereau et al. (EPO Application 0563485A1) which are incorporated herein by reference.
  • T-cells isolated from individuals can be exposed to the MART-1 protein or portions thereof, or MART-1 peptides or analogs thereof or MART-1 modified peptides or analogs thereof in vitro and then administered to a patient in need of such treatment in a therapeutically effective amount.
  • T-lymphocytes include but are not limited to, peripheral blood cells lymphocytes (PBL), lymph nodes, or tumor infiltrating lymphocytes (TIL).
  • PBL peripheral blood cells lymphocytes
  • TIL tumor infiltrating lymphocytes
  • lymphocytes can be isolated from the individual to be treated or from a donor by methods known in the art and cultured in vitro (Kawakami, Y. et al. (1989) J. Immunol. 142: 2453-3461).
  • Lymphocytes are cultured in media such as RPMI or RPMI 1640 or AIM V for 1-10 weeks. Viability is assessed by trypan blue dye exclusion assay.
  • the lymphocytes are exposed to all or part of the MART-1 protein for part or all of the culture duration.
  • the lymphocytes are exposed to the AAGIGILTV (SEQ ID NO: 4) peptide (presented in single letter code) at a concentration of about 1 to about 10 micrograms(ug)/ml per 10 7 cells for all or part of the duration of lymphocyte culture. After being sensitized to the peptide the T-lymphocytes are administered to the mammal in need of such treatment.
  • the modified MART-1 peptides shown in Table 14 may be exposed to lymphocytes.
  • these sensitized T-cells include but are not limited to, intravenously, intraperitoneally or intralesionally.
  • Parameters that may be assessed to determine the efficacy of these sensitized T-lymphocytes include, but are not limited to, production of immune cells in the mammal being treated or tumor regression. Conventional methods are used to assess these parameters. Such treatment can be given in conjunction with cytokines or gene modified cells (Rosenberg, S. A. et al. (1992) Human Gene Therapy, 3: 75-90; Rosenberg, S. A. et al. (1992) Human Gene Therapy, 3: 57-73).
  • the compositions can be used to prepare antibodies to MART-1 antigen, peptides or analogs thereof, or modified MART-1 peptides and analogs thereof.
  • the antibodies can be used directly as anti-melanoma agents.
  • a host animal is immunized using the MART-1 protein, peptides or analogs thereof, or modified peptides or analogs thereof as the immunogen and bound to a carrier as described above for vaccines.
  • the host serum or plasma is collected following an appropriate time interval to provide a composition comprising antibodies reactive with the immunogen.
  • the gamma globulin fraction or the IgG antibodies can be obtained, for example, by use of saturated ammonium sulfate or DEAE Sephadex, or other techniques known to those skilled in the art.
  • the antibodies are substantially free of many of the adverse side effects which may be associated with other anti-cancer agents such as chemotherapy.
  • the antibody compositions can be made even more compatible with the host system by minimizing potential adverse immune system responses. This is accomplished by removing all or a portion of the Fc portion of a foreign species antibody or using an antibody of the same species as the host animal, for example, the use of antibodies from human/human hybridomas.
  • Humanized antibodies i.e., nonimmunogenic in a human
  • Such chimeric antibodies may contain the reactive or antigen binding portion of an antibody from one species and the Fc portion of an antibody (nonimmunogenic) from a different species.
  • chimeric antibodies include but are not limited to, non-human mammal-human chimeras, rodent-human chimeras, murine-human and rat-human chimeras (Robinson et al., International Patent Application 184,187; Taniguchi M., European Patent Application 171,496; Morrison et al., European Patent Application 173,494; Neuberger et al., PCT Application WO 86/01533; Cabilly et al., 1987 Proc. Natl. Acad. Sci. USA 84:3439; Nishimura et al., 1987 Canc. Res. 47:999; Wood et al., 1985 Nature 314:446; Shaw et al., 1988 J. Natl. Cancer Inst. 80: 15553, all incorporated herein by reference).
  • Suitable “humanized” antibodies can be alternatively produced by CDR or CEA substitution (Jones et al., 1986 Nature 321:552; Verhoeyan et al., 1988 Science 239:1534; Siedleret al. 1988 J. Immunol. 141:4053, all incorporated herein by reference).
  • the antibodies or antigen binding fragments may also be produced by genetic engineering.
  • the technology for expression of both heavy and light cain genes in E. coli is the subject the following PCT patent applications; publication number WO 901443, WO901443, and WO 9014424 and in Huse et al., 1989 Science 246:1275-1281.
  • the antibodies can also be used as a means of enhancing the immune response.
  • the antibodies can be administered in amounts similar to those used for other therapeutic administrations of antibody.
  • pooled gamma globulin is administered at a range of about 1 mg to about 100 mg per patient.
  • antibodies reactive with the MART-1 antigen can be passively administered alone or in conjunction with other anti-cancer therapies to a mammal afflicted-with melanoma.
  • anti-cancer therapies include, but are not limited to, chemotherapy, radiation therapy, adoptive immunotherapy therapy with TIL.
  • anti MART-1 antigen antibodies can be induced by administering anti-idiotype antibodies as immunogens.
  • a purified anti-MART-1 antibody preparation prepared as described above is used to induce anti-idiotype antibody in a host animal.
  • the composition is administered to the host animal in a suitable diluent. Following administration, usually repeated administration, the host produces anti-idiotype antibody.
  • antibodies produced by the same species as the host animal can be used or the Fc region of the administered antibodies can be removed.
  • serum or plasma is removed to provide an antibody composition.
  • composition can be purified as described above for anti-MART-1 antibodies, or by affinity chromatography using anti-MART-1 antibodies bound to the affinity matrix.
  • anti-idiotype antibodies produced are similar in conformation to the authentic MART-1-antigen and may be used to prepare an MART-1 melanoma antigen vaccine rather than using the MART-1 protein, peptides analogs or portions thereof.
  • the manner of injecting the antibody is the same as for vaccination purposes, namely intramuscularly, intraperitoneally, subcutaneously, interlesionally, or the like in an effective concentration in a physiologically suitable diluent with or without adjuvant.
  • One-or more booster injections may be desirable.
  • the MART-1 derived proteins or peptides or modified peptides of this invention are also intended for use in producing antiserum designed for pre- or post-disease prophylaxis.
  • the MART-1 antigen, peptides or analogs thereof, or modified MART-1 peptides or analogs thereof is formulated with a suitable adjuvant and administered by injection to human volunteers, according to known methods for producing human antisera.
  • Antibody response to the injected proteins is monitored, during a several-week period following immunization, by periodic serum sampling to detect the presence of anti-MART-1 serum antibodies, using an immunoassay as described herein.
  • the antiserum from immunized individuals may be administered as a prophylactic measure for individuals who are at risk of developing melanoma.
  • the antiserum is also useful in treating an individual afflicted with melanoma for post-disease prophylaxis.
  • Monoclonal anti-MART-1 antibodies or anti-idiotype antibodies can be produced as follows. The spleen or lymphocytes from an immunized animal are removed and immortalized or used to prepare hybridomas by methods known to those skilled in the art. (Goding, J. W. 1983. Monoclonal Antibodies: Principles and Practice, Pladermic Press, Inc., NY, N.Y., pp. 56-97). To produce a human-human hybridoma, a human lymphocyte donor is selected.
  • a donor known to have a melanoma carrying the MART-1 antigen may serve as a suitable lymphocyte donor.
  • Lymphocytes can be isolated from a peripheral blood sample or spleen cells may be used if the donor is subject to splenectomy.
  • Epstein-Barr virus (EBV) can be used to immortalize human lymphocytes or a human fusion partner can be used to produce human-human hybridomas.
  • Primary in vitro immunization with peptides can also be used in the generation of human monoclonal antibodies.
  • MART-1 peptides examples include AAGIGILTV (SEQ ID NO: 4), EAAGIGILTV (SEQ ID NO: 17) and AAGIGILTVI (SEQ ID NO: 18) (peptides are presented in single letter amino acid code). Most preferably AAGIGILTV (SEQ ID NO: 4) is used as the immunogen. Alternatively, peptides derived from the MART-1 amino acid sequence and modified to enhance binding of the peptide to a MHC Class I molecule may also be used. By way of example the modified peptides shown in Table 14 may be used as the immunogen.
  • Antibodies secreted by the immortalized cells are screened to determine the clones that secrete antibodies of the desired specificity.
  • the antibodies For monoclonal MART-1 antigen or peptide antibodies, the antibodies must bind to MART-1 antigen or peptide.
  • the antibodies For monoclonal anti-idiotype antibodies, the antibodies must bind to anti-MART-1 antibodies.
  • Cells producing antibodies of the desired specificity are selected.
  • the antibodies or chimeric antibodies described herein may also be coupled to toxin molecules radio-isotopes and drugs by conventional methods (Vitetta et al. (1991) in “Biologic Therapy of Cancer” De Vita V T, Hellman S., Rosenberg, S. A. (eds) J.B. Lippincott Co. Philadelphia; Larson, S. M. et al. (1991) in “Biological Therapy of Cancer” De Vita V. T., Hellman S., Rosenberg, S. A. (eds) J.B. Lippincott Co., Philadelphia).
  • Examples of toxins to which the antibodies may be coupled to include, but are not limited to, ricin or diphtheria toxin.
  • drugs or chemotherapeutic agents include, but are not limited to, cyclophosphamide or doxorubcin.
  • radioisotopes include, but are not limited to, 131 I, Antibodies covalently conjugated to the aforementioned agents can be used in cancer immunotherapy for treating melanoma.
  • Local administration to the afflicted site may be accomplished through means known in the art, including, but not limited to, topical application, injection, and implantation of a porous device containing cells recombinantly expressing the infusion, implantation of a porous device in which the MART-1 antibodies or chimeric antibodies, antibodies coupled to toxins, drugs or radiolabels or portions thereof are contained.
  • the above described antibodies and antigen binding fragments thereof may be supplied in kit form alone, or as a pharmaceutical composition for in vivo use.
  • the antibodies may be used for therapeutic uses, diagnostic use in immunoassays or as an immunoaffinity agent to purify the MART-1 protein or peptides as described herein.
  • the present invention also provides a substantially purified and isolated nucleic acid sequence, designated c(complementary)DNA25 ( FIG. 4 ; SEQ. ID. NO. 26) which encodes a second melanoma recognized by tumor infiltrating lymphocytes.
  • the TIL which recognize the melanoma antigen encoded by cDNA25 are associated with in vivo tumor rejection.
  • the TIL recognized the melanoma antigen encoded by cDNA25 in the context of HLA-A2.
  • Comparison of the cDNA25 nucleic acid sequence FIG. 4 ; SEQ.
  • Previously identified sequences for gp100 include gp100 (GenBank Accession No. M32295; also designated gp95), Pmel 17 (GenBank Accession No. M77348; Kwon et al., (1991) Proc. Natl. Acad. Sciences (USA) 88:9228-9232) and ME20 (Maresh et al. (1994) DNA and Cell Biology 13:87-95).
  • the cDNA25 sequence provided herein differs from the previously reported gp100 sequence in Genbank (Genbank Accession No. M32295) by two nucleotides, from the Pmel 17 sequence (Kwon et al. (1991) Proc. Natl. Acad. Sciences (USA) 88: 9228-9232) by three nucleotides and a twenty one base pair deletion, and from the ME20 sequence (Maresh et al. (1994) DNA and Cell Biology 13:87-95) by a single nucleotide difference.
  • the protein encoded by cDNA25 differs from the gp100 in GenBank (GenBank Accession #M32295) by one amino acid at position 162, by a two amino acid difference at positions 162 and 274, compared to Pmel 17 and did not contain 7 amino acids that existed in Pmel 17 at positions 588-594. Therefore, cDNA25 appears to encode for a different form of the gene for gp100.
  • the differences between the cDNA25 nucleic acid sequence ( FIG. 4 ; SEQ. ID. NO. 26) and amino acid sequence ( FIG. 5A ; SEQ. ID. NO. 27) and previously reported gp100 sequences may be due to polymorphisms, allelic variations, or to mutations within the tumor. Experiments with mouse tumors have shown that new antigens recognized by T-cells can result from point mutation in the coding region of the inactive gene (Boon, T (1992) Advances in Cancer Research 58:177-210).
  • This invention also provides immunogenic peptides derived from the gp100 protein sequence provided herein. ( FIG. 5A ; SEQ. ID. NO. 27). These immunogenic peptides represent the antigenic portions of the gp100 protein ( FIG. 5A ; SEQ. ID. NO. 27) recognized by TIL. Examples of immunogenic peptides include, but are not limited to, LLDGTATLRL (peptide G10-4; SEQ ID NO: 33; FIG. 4 , nucleotides 1407-1436), VLYRYGSFSV (peptide G10-5; SEQ ID NO: 34; FIG. 4 nucleotides 1464-1493), ALDGGNKHFL (peptide G10-22; SEQ ID NO: 35; FIG.
  • FIG. 4 nucleotides 708-737), VLKRCLLHL (peptide G9-19 SEQ ID NO: 36; FIG. 4 , nucleotides 48-74), VLPSPACQLV (peptide G10-8; SEQ ID NO: 37; FIG. 4 nucleotides 1668 to 1797) SLADTNSLAV (peptide G10-9; SEQ ID NO: 38; FIG. 4 nucleic acids 1746 to 1775) SVSVSQLRA (peptide G9-216; SEQ ID NO:39; FIG. 4 , nucleotides 684 to 710) YLEPGPVTA (peptide G9-280; SEQ ID NO:40; FIG.
  • nucleotides 876-902) or LNVSLADTN (peptide G10-400; SEQ ID NO:41; FIG. 4 ; nucleotides 1736-1764) KTWGQYWQV (peptide G9 154 ; SEQ ID NO:46; FIG. 5A ; amino acids 154 to 162), KTWGQYWQVL (peptide G10 154 SEQ ID NO:47; FIG. 5A ; amino acids 154 to 163), ITDQVPFSV (peptide G9 209 , SEQ ID NO:48; FIG. 5A ; amino acids 209 to 217) and TITDQVPFSV (peptide G10 208 ; SEQ ID NO: 49; FIG.
  • This invention further includes analogs of these immunogenic peptides derived from the gp100 amino acid sequence ( FIG. 5A ; SEQ ID NO: 27).
  • the term analog includes any peptide which displays the functional aspects of these immunogenic peptides.
  • the term analog also includes conservative substitution or chemical derivative of the peptides as described above.
  • These immunogenic peptides may be synthetically or recombinantly produced in the same manner or fashion as described above for MART-1.
  • immunogenic peptides derived from the gp100 sequence are modified to increase immunogenicity by enhancing the binding of the peptide to MHC molecule with which the peptide is associated when presented to T-cells.
  • modifications may include the substitution, deletion or addition, of one or more amino acids within the immunogenic peptide sequence, or insertion of amino acids within the given immunogenic peptide sequence or derivitization of existing amino acids within the given immunogenic peptide sequence or mutation of the amino acids within the given immunogenic peptide sequence.
  • at least one amino acid is substituted or replaced in the given immunogenic peptide sequence.
  • Any amino acid composing the given immunogenic peptide sequence may be modified in accordance with this invention. Any amino acid may be used to substitute or replace a given amino acid within the immunogenic peptide sequence. Modification may occur at any amino acid position within the immunogenic gp100 peptide. Modified gp100 peptides is intended to include any modified immunogenic gp100 peptide exhibiting enhanced binding with the MHC molecule with which it is associated when presented to the T-cell.
  • peptides recognized by T cells in the context of HLA-A2 are 9 to 10 amino acids in length.
  • the second position and last position in the peptide are hydrophobic amino acids preferably aliphatic hydrophobic amino acids.
  • the second position may be any aliphatic hydrophobic amino acid such as, but not limited to, leucine, methionine, isoleucine, valine, threonine, glycine or alanine.
  • the last position of the peptide may be any aliphatic hydrophobic amino acid, such as but not limited to valine, leucine, alanine, leucine, isoleucine, glycine, methionine, valine, or threonine.
  • the first and third positions of the immunogenic peptide may be substituted or replaced with any amino acid, preferably, hydrophobic aliphatic amino acids, or aromatic amino acids.
  • amino acids that may be used at the first or third position of the peptide include, but are not limited to, alanine, leucine, lysine, isoleucine, glycine, methionine, valine, threonine, tryptophan, phenylalanine, serine, lysine or tyrosine.
  • Examples of gp100 peptides that may be modified in accordance with the present embodiment include, but is not limited to LLDGTATLRL (peptide G10-4; SEQ ID NO: 33; FIG. 4 , nucleotides 1407-1436), VLYRYGSFSV (peptide G10-5; SEQ ID NO: 34; FIG. 4 nucleotides 1464-1493), ALDGGNKHFL (peptide G10-22; SEQ ID NO: 35; FIG. 4 , nucleotides 708-737), VLKRCLLHL (peptide G9-19 SEQ ID NO: 36; FIG. 4 , nucleotides 48-74), VLPSPACQLV (peptide G10-8; SEQ ID NO: 37; FIG.
  • SLADTNSLAV (peptide G10-9; SEQ ID NO: 38; FIG. 4 nucleic acids 1746 to 1775) SVSVSQLRA (peptide G10-216; SEQ ID NO:39; FIG. 4 , nucleotides 684 to 710) YLEPGPVTA (peptide G9-280; SEQ ID NO:40; FIG. 4 ; nucleotides 876-902) or LNVSLADTN (peptide G10-400; SEQ ID NO:41; FIG. 4 ; nucleotides 1736-1764) KTWGQYWQV (peptide G9 154 ; SEQ ID NO:46; FIG.
  • modified gp100 peptides derived from the immunogenic gp100 peptide KTWGQYWQV may have the formula X 1 X 2 X 3 GQYWQX 4 wherein:
  • X 1 may be any amino acid, preferably any hydrophobic aliphatic amino acid, or aromatic amino acid.
  • amino acids that may be used include, but are not limited to, alanine, leucine, lysine, isoleucine, glycine, methionine, valine, threonine, tryptophan, phenylalanine, lysine or serine, aspartic acid or tyrosine;
  • X 2 may be any hydrophobic amino acid, preferably any aliphatic hydrophobic amino acid.
  • amino acids that may be used include, but are not limited to, leucine, methionine, isoleucine, alanine, threonine, glycine, or valine. Most preferably leucine, methionine or isoleucine.
  • X 3 may be any amino acid, preferably any hydrophobic aliphatic amino acid or aromatic amino acid.
  • amino acids that may be used include, but are not limited to, alanine, leucine, lysine, isoleucine, glycine, methianine, valine, threonine, tryptophan, phenylalanine, serine, lysine, or tyrosine;
  • X 4 is any hydrophobic amino acid, preferably an aliphatic hydrophobic amino acid.
  • amino acids that may be used include, but are not limited to, valine, leucine, isoleucine, methionine, alanine, threonine, or glycine.
  • modified peptides are shown in Table 15.
  • a preferred modified peptide is KIWGQYWQV (G9-154-2I).
  • immunogenic gp100 ITDQVPFSV (G9-209; SEQ ID NO:48) may be modified, such modified peptides may have the general formula X 1 X 2 X 3 QVPFSX 4 wherein:
  • X 1 may be any amino acid, preferably any hydrophobic aliphatic amino acid, or aromatic amino acid.
  • amino acids that may be used include, but are not limited to, leucine, methionine, alanine, isoleucine, valine, threanine, glycine, lysine, phenylalanine, tryptophan, or tyrosine, aspartic acid or serine;
  • X 2 is any hydrophobic amino acid, preferably a hydrophobic aliphatic amino acid.
  • amino acids that may be used include, but are not limited to, leucine, methionine, alanine, isoleucine, valine, threanine, or glycine;
  • X 3 may be any amino acid, preferably any hydrophobic, aliphatic amino acid or aromatic amino acid.
  • amino acids that may be used include, but are not limited to, leucine, methionine, alanine, isoleucine, valine, threonine, glycine, lysine, phenylalanine, tryptophan, tyrosine, aspartic acid or serine;
  • X 4 may be any hydrophobic amino acid, preferably any hydrophobic aliphatic amino acid.
  • amino acids that may be used include, but are not limited to, leucine, methionine, alanine, isoleucine, valine, or threonine;
  • modified peptides derived from ITDQVPFSV are shown in Table 16.
  • the peptide FLDQVPFSV peptide G9-209-1F2L is used.
  • modified gp100 peptides derived from the immunogenic gp100 peptide YLEPGPVTA may also be modified to enhance binding to MHC Class I molecules, preferably HLA-A2 or subtypes thereof.
  • the modified peptides may have the general formula X 1 X 2 X 3 PGPVTX 4 wherein:
  • X 1 may be any amino acid, preferably a hydrophobic aliphatic amino acid or aromatic amino acid.
  • amino acids that may be used include, but are not limited to, leucine, methionine, alanine, isoleucine, valine, threanine, glycine, lysine, phenylalanine, tryptophan, or tyrosine, aspartic acid or serine;
  • X 3 may be any amino acid, preferably any hydrophobic aliphatic amino acid, or aromatic amino acid.
  • amino acids that may be used include, but are not limited to, leucine, methionine, alanine, isoleucine, valine, threanine, glycine, lysine, phenylalanine, tryptophan, tyrosine, aspartic acid or serine;
  • X 4 may be any hydrophobic amino acid preferably an aliphatic hydrophobic amino acid.
  • amino acids that may be used include but are not limited to, leucine, methionine, alanine, isoleucine, valine, threonine, or glycine.
  • modified peptides derived from YLEPGPVTA (G9-280; SEQ ID NO:40) are shown in Table 17.
  • a preferred modified peptide is YLEPGPVT (G9-280-9V).
  • This invention further includes analogs of these modified peptides derived from the gp-100 sequence ( FIG. 5A ; SEQ ID NO:27).
  • the term analog is intended to include any peptide which displays the functional aspects of these modified peptides as described above.
  • These modified peptides may be synthetically or recombinantly provided by conventional methods.
  • the immunogenic peptides (SEQ ID NO: 33 to 38 and 46 to 49) derived from the gp100 amino acid sequence (SEQ ID NO: 27) or modified gp100 peptides as shown in Tables 15-17 or analogs thereof, may be used as a vaccine either therapeutically or prophylactically.
  • the vaccine is provided in advance of any evidence of melanoma.
  • the prophylactic administration of these peptides should serve to prevent or attenuate melanoma in a mammal.
  • mammals preferably humans, at high risk for melanoma are prophylactically treated with these vaccines.
  • the vaccine may be provided therapeutically to enhance the patients own immune response to the tumor antigen prescribed on the melanoma or metastatic melanoma.
  • the vaccine which acts as an immunogen, may be a cell, cell lysate from cells transfected with a recombinant expression vector carrying a nucleic acid sequences encoding gp100 immunogenic peptide or a culture supernatant containing the expressed protein. Expression vectors into which nucleic acid sequences encoding these immunogenic peptides may be introduced are the same as those described above for MART-1.
  • the immunogen is a partially or substantially purified recombinant gp100 peptide or analog thereof.
  • the immunogen While it is possible for the immunogen to be administered in a pure or substantially pure form, it is preferable to present it as pharmaceutical compositions, formulations or preparations as described above for MART-1. Vaccination can be conducted by conventional methods previously described above for MART-1.
  • the gp100 immunogenic peptides and nucleic acids sequences encoding them may be used in bioassays, or to generate antibodies in the same manner or fashion as described above for MART-1.
  • multivalent vaccines against one or more melanoma antigens are provided.
  • Such multivalent vaccines may comprise all or part of the MART-1 protein peptides or modified peptides or gp100 peptides or modified peptides or combinations thereof.
  • this invention also provides a method of identifying genes encoding melanoma antigens utilizing cDNA expression cloning using tumors infiltrating lymphocytes isolated (TIL) from the tumor of patients afflicted with melanoma.
  • TIL tumor infiltrating lymphocytes isolated
  • the method comprises the following steps: (a) isolating tumor infiltrating lymphocytes from the tumor of a mammal afflicted with melanoma; (b) introducing a melanoma cDNA library into a mammalian cell line; (c) exposing said mammalian cells to said TIL; (d) screening for expression of an antigen encoded by said cDNA in said mammalian cells recognized by said TIL; and (e) isolating said cDNA corresponding to said antigen.
  • the tumor infiltrating lymphocytes in step (a) may be isolated from patients afflicted with melanoma including, but not limited to, the melanoma lesion, subcutaneous tissue or visceral organs.
  • Examples of cells that may be used to prepare the cDNA library used in step (b), include, but are not limited to, fresh or cultured melanoma cells.
  • the cDNA library is introduced into mammalian cells not expressing melanoma antigens. If non human mammalian cells or human cells not expressing the desired HLA haplotype for recognition by the TIL are used in step (b), such cells can be cotransfected with an HLA gene as described below.
  • Examples of cells which can be used in step (b) include but are not limited to, tumor cell lines, such as breast cancer cell line MDA 231 (ATCC #HTB26), or COS 7 cells (ATCC #CRL 1651).
  • MHC genes which can be used include, but are not limited to, HLA-A, HLA-B, and HLA-C genes, preferably HLA-A2 and subtypes thereof (Zemmour, J. et al. (1992) Tissue Antigens 40:221-228).
  • the appropriate MHC gene to be used is determined by the haplotype of the tumor cells which were the source for the cDNA library. Standard methods can be used to determine the haplotype recognized by the TIL isolated ( ASHI Laboratory Manual (2nd Edition 1990).
  • Examples of how to evaluate recognition of the cells containing the cDNA clone expressing an antigen recognized by the TIL includes, but is not limited to, ⁇ -interferon assays, TNF secretion (Van de Bruggen et al., (1991) Science 254:1643-1647) or lysis of cells transfected with cDNA encoding for the recognized antigen. Such assays are performed by conventional methods known to one skilled in the art. Melanoma antigens can be isolated by or rescued by PCR using primer specific to flanking site of vector containing the cDNA. Examples of how to isolate the cDNA corresponding to the antigen recognized by the TIL include, but are not limited to, PCR.
  • a method for assessing the immunogenicity of peptides derived from the predicted amino acid sequences of the MART-1 protein ( FIG. 1 ; SEQ ID NO: 2) or a gp100 protein ( FIG. 5A ; SEQ ID NO: 27).
  • the method comprises the steps of: (a) preparing a plurality of peptides based on the MART-1 ( FIG. 1 ; SEQ ID NO: 2) or gp100 ( FIG.
  • step (b) amino acid sequence
  • step (b) incubating at least one of said peptides with a mammalian cell line
  • step (c) exposing said mammalian cells incubated with said peptide to tumor infiltrating lymphocytes (TIL); and (d) screening for recognition of TIL with said cells incubated with said peptide.
  • TIL tumor infiltrating lymphocytes
  • peptides of about 25 to 5 amino acids be used, more preferably 20 to 10 amino acids and most preferably 9-10 amino acids.
  • cells that may be used in step (b) include, but are not limited to, T2-cells, (Cerundolo, V. et al.
  • T cells recognize antigen complexed with MHC Class 1 molecules.
  • the MHC locus in all mammalian species contains numerous genes and is highly polymorphic. Different MHC molecules or haplotypes types bind different antigens.
  • the HLA complex contains the HLA-A, HLA-B and HLA-C gene loci which encode class I molecules. Lymphocytes will recognize tumor antigens on the context of HLA Class 1 molecule. If the cells containing the recombinant MART-1 expression vector are to be screened by the TIL but are not human cells, such as COS cells, or do not express a desired haplotype an expression vector containing an MHC Class 1 gene may also be introduced into the cells. (See Example 1) This, represents yet another alternative embodiment of the invention.
  • MHC Class I genes that may be used include, but are not limited to, HLA-A, HLA-B and HLA-C genes.
  • MHC specificities or restriction types include, but is not limited to HLA-A1, HLA-A2, such as the HLA-A2.1 subtype, or HLA-A24 (Zemmour, J. et al. (1992) Tissue Antigens 40:221-228). Most preferred is the HLA-A2.1 gene.
  • compositions and therapeutic applications are also intended to be encompassed bit the compositions and therapeutic applications described herein.
  • CTL Cytotoxic T Lymphocytes
  • ⁇ 54 CTL were generated from excised tumor specimens by culturing a suspension of cells with 6000 IU/ml of IL-2 (Cetus-Oncology Division, Chiron Corp. Emeryville, Calif.) for 30-70 days as described in Kawakami, Y., et al. (1988) J. Exp. Med. 168:2183-2191.
  • TIL501 and TIL1235 were predominantly CD8 + and were derived from the tumor specimens of patients with advanced metastatic melanoma.
  • TIL501.A42 The CD8 + T cell clone, TIL501.A42, was established by limiting dilution methods and cultured with 120 IU/ml of IL-2 plus irradiated (once a week for 4 to 6 times) autologous tumor cells.
  • NHEM483, NHEM493, NHEM527, NHEM529, NHEM530, NHEM533, NHEM616 and NHEM680 were purchased from Clonetics, San Diego, Calif., FM725, FM801, FM902 were provided by M. Herlyn, Wistar Institute, Philadelphia Pa., HA002 was provided by R. Halaban, Yale university, New Haven, Conn. and cultured in melanocyte growth medium (MGM, Clonetics).
  • Ewing's sarcoma cell lines, TC71, RD-ES, 6647 were provided by M. Tsokos, NIH, Bethesda, Md.
  • the neuroblastoma cell line, SK—N-AS was provided by O. M. El Badry, NIH, Bethesda, Md.
  • the plasmacytoma cell line, HMY-C1R and the M1 fibroblast cell line were provided by W. Biddison, NIH, Bethesda, Md. Kidney epithelial cells, KAM, WLC were provided by D. J. Hazen-Martin and D. A. Sens, Medical University of South Carolina, Charleston, S.C.
  • the monkey kidney cell line, COS7 was provided by W. Leonard, NIH, Bethesda, Md.
  • 51 Cr release assays were performed as described in Kawakami, Y., et al., (1988) J. Exp. Med. 168:2183-2191. Briefly, 5000 target cells labeled with 51 Cr were mixed with varying numbers of effector cells and incubated for 5 hours (h). Then supernatants were collected, radioactivity was measured and the percent specific lysis was calculated.
  • a cDNA library was constructed from poly A RNA from the HLA-A2 + melanoma cell line, 501mel as described in (Miki, T., et al., (1989) Gene; 83:137-146 Miki et al. (1991) Proc. Nat. Acad. Sci. USA 88:5167-5171). Briefly, first strand cDNA was synthesized with a linker primer GGACAGGCCGAGGCGGCC(T) 40 followed by second strand cDNA synthesis. After treatment with T4 DNA ligase, an SfiI adaptor consisting of two oligonucleotides, CCAATCGCGACC and GGTCGCGATTGGTAA was ligated to the end of the cDNA.
  • the cDNA was digested with SfiI and the digested fragment was isolated by passing through a spun column.
  • the cDNA was then mixed with bacteriophage ⁇ pCEV27 (Miki, T. et al., (1991) Proc. Natl. Acad. Science (USA) 88: 5167-5771) vector arms prepared by SfiI digestion and in vitro packaging was performed.
  • PCR Polymerase chain reaction
  • the pcDNA3 containing the cloned genes were transiently transfected into the COS7 cell line by the DEAE dextran method (Seed, B. and Aruffo, A. (1987) Proc. Natl. Acad. Sci. (USA) 84:3365-3369). Briefly, 3 ⁇ 10 5 cells per well in 6 well plates were incubated at 37° C.
  • the ability of the transfected COS7 or 293 cells to mediate IFN- ⁇ release from TIL was assessed.
  • the expression of the HLA-A2 gene was tested by flow cytometry. Stable transfectants were made by the calcium-phosphate method and individual colonies and pooled transfectants were tested for reactivity to TIL by cytotoxicity and IFN- ⁇ release assays.
  • DNA sequencing of the cloned genes was performed by dideoxy chain termination method with dGTP and 7-deaza-dGTP.
  • DNA and protein sequences were analyzed by the GCG program with GeneBank, and EMBL Data Library nucleotide databases and SWISS-PROT, PIR, GenPept, Brookhaven Protein Data Bank protein databases.
  • Hybridization with the probe was performed according to the QuikHyb protocol (Stratagene) at 68 C for 2-16 h. Membranes were washed two times with 2 ⁇ SSC/0.1% SDS at 60 C for 15 minutes (min) and once with 0.1 ⁇ SSC at 60 C at for 30 min and then autoradiography was performed.
  • TIL lines were established from HLA-A2 + melanoma patients and tested for lysis of melanoma cell lines from HLA-A2 + and HLA-A2 ⁇ patients.
  • HLA typing of patients is performed by conventional HLA typing techniques.
  • HLA-A2 was selected because it is the most frequently expressed class I MHC antigen (about 50% of individuals) and has been shown to be a dominant restriction element for the recognition of melanoma antigens (Crowley, N. J., et al. (1991), J. Immunol. 146:1692-1694).
  • TIL501, TIL1235 and TIL1200 exhibited specific recognition of shared melanoma antigens in an HLA-A2 restricted fashion.
  • TIL501.A42 was a T cell clone established from TIL501 by limiting dilution. These TIL caused lysis or released cytokines including IFN- ⁇ , TNF ⁇ and GM-CSF when cocultured with a variety of HLA-A2 + melanoma or melanocyte cell lines but not HLA-A2 ⁇ melanoma lines or HLA-A2 + non-melanoma cell lines including the breast cancer cell line, MDA 231. Two representative experiments are shown in Table 1. Thus, these CTL seemed to recognize a non-mutated peptide derived from a melanocyte lineage specific antigen.
  • a cDNA library from the HLA-A2 + 501mel melanoma cell line was transfected into two highly transfectable HLA-A2.1 + cancer cell lines, MDA231 and A375. These cell lines were not lysed by melanoma specific TIL but were lysed by HLA-A2 restricted influenza M1 specific CTL after incubation with the M1 58-66 peptide (GILGFVFTL; single letter code) derived the influenze matrix protein or infection with a recombinant vaccinia virus containing the M1 gene (data not shown).
  • GILGFVFTL single letter code
  • these cell lines exhibited normal antigen processing and presenting ability but were not lysed by these melanoma-specific TIL because of the lack of expression of the relevant melanoma antigens.
  • G418 After selection with G418, approximately 6700 transfected clones from each cell line were isolated and grown in microplates. Using the IFN- ⁇ release assay, 21 MDA231 and 27 A375 positive clones were isolated and rescreened. Of these clones, eight MDA231 and seven A375 clones were positive in a second screening assay.
  • PCR using genomic DNA from these positive transfectants was performed with SP6 and T7 primers flanking the insert genes.
  • the 1.6 Kb band detected by Northern blot analysis with the cDNA 22 probe suggested that this fragment was a full length cDNA.
  • Transient transfection of the expression vector pcDNA3 containing the cDNA from clones 22 or 23 into either COS7 or 293 cells along with the HLA-A2.1 gene conferred reactivity to TIL1235 and TIL501.A42 as demonstrated by the specific release of IFN- ⁇ (Table 2, Experiments 1 and 2). Stable transfection of these cDNA fragments into MDA 231 or A375mel cell lines also conferred reactivity to TIL1235 and TIL501.42 (Table 2, Experiment 3). TIL501.A42 could lyse MDA231 stably transfected with cDNA 22 (data not shown).
  • TIL 501.A42 and TIL 1235 for Melanomas and Melanocytes
  • TIL586 is class I MHC restricted melanoma specific TIL, not restricted by HLA-A2.
  • the cDNA sequence of clones 22 and 23 were identical except at a single base that was believed to be a change introduced by PCR. Two other independently amplified fragments were also sequenced to clarify this region and the consensus sequence is shown in FIG. 1 .
  • the longest open reading frame in this gene consists of 354 bases corresponding to a 118 amino acid protein of 13 kd. This sequence did not show significant similarity to any complete nucleotide or protein sequences in established databases.
  • Amino acids 27-47 consist of a hydrophobic region that may contain the HLA-A2 binding peptides (Falk, K., et al. (1991), Nature 351:290-296; Hunt, D. F., et al.
  • MART-1 antigen Melanoma Antigen Recognized by T cells-1.
  • HLA-A2 restricted TIL lines generated nine recognized MART-1, and four recognized a form of gp100 isolated and described herein (see Example 3) and none appeared to recognize MAGE-1 (Zakut, R., et al. (1993), Cancer Res 53:5-8.; data not shown).
  • TIL501.A42 and TIL 1235 When Cultured with HLA-A2 + Cell Lines Transfected with the Gene 22 or 23 TIL501.A42 TIL1235 Stimulator Cell Transfected Gene HLA-A2 pg/ml Exp. 1 501mel none + 1009* 1076 397mel none ⁇ ⁇ 50 ⁇ 50 COS7 none ⁇ ⁇ 50 ⁇ 50 COS7 HLA-A2.1 + ⁇ 50 ⁇ 50 COS7 22 ⁇ ⁇ 50 ⁇ 50 COS7 HLA-A2.1+22 + 771 1049 Exp.
  • IFN- ⁇ was secreted only when TIL were incubated with HLA-A2 + cell lines transfected with the cDNA 22 or 23.
  • TIL 501.A42 which recognized the MART-1 Ag lysed 13 of 17 (76%) HLA-A2 + melanoma cell lines tested (data not shown).
  • MART-1 is a previously undescribed antigen expressed on melanocyte lineage cells from skin and retina that is also expressed on melanoma cells.
  • MART-1 FIG. 1 ; SEQ ID NO. 1
  • gp100 a gene encoding melanoma antigens
  • HLA-A2.1 restricted TIL a gene encoding melanoma antigen.
  • the MART-1 antigen is a 118 amino acid protein of approximately 13 kd. Neither the gene nor the amino acid sequence for MART-1 have been previously described.
  • MART-1 RNA was expressed in 11 of 14 HLA-A2.1 positive or negative melanoma lines, and 7 of 7 melanocyte lines. With the exception of retinal tissue no MART-1 expression was found on any normal tissue tested, T-cell lines, B-cell lines, kidney epithelial lines, a fibroblast line or 19 tumor cell lines from cancers of the colon, breast, brain, kidney, lung or bone.
  • MAGE-1 Another melanoma antigen, MAGE-1, has been described that is recognized by T cells derived from peripheral blood lymphocytes following repetitive in vivo or in vitro immunization (Van Der Bruggen, et al. (1991), Science 254:1643-1647).
  • TIL with anti melanocyte-melanoma reactivities have been administered to patients with advanced melanoma (Rousenberg, S. A., et al. (1988), N Engl J Med 319:1076-1680; Rosenberg S. A., J. Clin. Oncol. 10:180-199) and although sporadic vitiligo has been seen in these patients, no adverse ophthalmologic effects related to the possible expression of these melanocyte antigens on retinal cells has been observed.
  • HLA-A2 is present in about 50% of individuals and the HLA-A2 restricted MART-1 antigen also appears to be widely expressed on melanomas, immunization with the MART-1 antigen may be particularly useful for the development of active immunotherapies.
  • cDNA encoding tyrosinase related protein(gp75) in pCEV27 plasmid was isolated from 501mel melanoma cDNA library using a probe generated by PCR based on the reported sequence of gp75 (Cohen et al. (1980) Nucleic Acids Research 18:2807).
  • Peptides were synthesized by a solid phase method using a Gilson AMS 422 multiple peptide synthesizer.
  • the peptides were purified by HPLC on a Vydac C-4 column with 0.05% TFA/water-acetonitrile.
  • TIL lysis of T2 cell lines preincubated for 2h with each peptide was measured using a 51 Cr release cytotoxicity assay.
  • HLA-A2 restricted melanoma specific CTL lines and a clone, A42 were established from lymphocytes infiltrating into tumors of 10 melanoma patients.
  • TIL recognized autologous and most. allogeneic fresh or cultured melanoma cells expressing HLA-A2, but did not recognize HLA-A2 melanomas or HLA-A2 + non-melanoma cell lines (Kawakami et al. (1992) J. Immunol 148:638). They also recognized HLA-A + normal cultured melanocytes derived from neonatal skin (see Example 1 and Kawakami, Y. et al. (1993), J. Immunotherapy 14:88). Thus, these TIL recognized non-mutated self-peptides derived from proteins expressed in melanoma and melanocytes in association with HLA-A2.
  • TIL1200 which is a relatively oligoclonal CTL line (Shilyansky, J. et al., (1994) Proc. Natl. Acad. Sci. (USA) 91:2829) did not respond to this COS transfectant.
  • TIL 620, 660, 1143, 1200
  • TIL1200 secreted large amounts of IFN- ⁇ compared to TIL620, 660, and 1143, suggesting that only a small subset of T-cells in these latter 3 TIL lines recognized gp100. None of these TIL recognized gp75 using this assay.
  • MART-1 is a common melanoma antigen recognized by most HLA-A2 restricted TIL derived from melanoma patients.
  • peptides were selected based on the known peptide binding motifs to HLA-A2.1 (Falk, K. et al., (1993) Nature, 351:290; Hunt, D. F. et al. (1992), Science, 255:1261; Ruppert, J. et al., (1993) Cell 74:929), synthesized (>90% purity) and screened by testing lysis of the HLA-A2.1 + T2 cell line by TIL after incubation of the T2 line with each peptide (Table 5).
  • the T2 cells (Cerundolo, V. et al.;
  • TIL1200 secreted IFN- ⁇ when cultured with COS7 cotransfected with cDNAs encoding MART-1 and HLA-A2.1.
  • TIL620, 660, 1143 and 1200 secreted IFN- ⁇ when cultured with COS7 cotransfected with the cDNAs encoding gp100 and HLA-A2.1. (1990) Nature 345: 449-452) cell line was lysed well by all 4 HLA-A2 restricted melanoma specific TIL tested when preincubated with either peptides M9-2, M10-3, or M10-4.
  • Both 10 amino acid peptides, M10-3 and M10-4 contain the M9-2 sequence, with M10-3 having an additional glutamic acid at its N-terminus and M10-4 having an extra isoleucine at its C-terminal end. These peptides are located in a hydrophobic putative transmembrane domain in MART-1. The same lysis was observed when other HLA-A2 + cells incubated with these peptides were used as targets including the K4B (provided by Dr. William Biddson, NIH; Storkus. W et al. (1993) J. of Immunology 151:3719-3727) and 501EBVB Epstein-Barr virus transformed B cells (Topalian et al. (1989) J.
  • the peptides, M9-1, M9-2, M9-3, M10-2, M10-3, M10-4 and M10-5 were further purified and titrated in order to evaluate their relative ability to sensitize T2 cells to lysis by MART-1 reactive TIL1235 or T cell clone A42 ( FIG. 2 ).
  • the purified peptides M9-2, M10-3 and M10-4 were required in minimum concentrations of 1 ng/ml, 10 ng/ml an 1000 ng/ml, respectively.
  • the purified M10-4 was not recognized by TIL clone A42 even at 10 ug/ml as shown in FIG. 2 .
  • M9-1, M9-3, M10-2, and M10-5 peptides were not recognized by either A42 or TIL1235.
  • TIL620 and TIL1088 demonstrated low level of non-specific lysis of T2 cells without peptides or after the addition of irrelevant peptides, but showed significant increase of lysis of T2 cells preincubated with M9-2, M10-3, and M10-5 peptides.
  • the recognition of M10-4 differed among the TIL, but was similar to the different reactivity to M10-4 by the T-cell clone A42 or the T-cell line TIL1235 ( FIG. 2 ). Higher concentrations (1 ug/ml) of M10-4 were required for lysis than were required for M9-2 or M10-3.
  • TIL and clone A42 also secreted cytokines including IFN- ⁇ , GM-CSF and TNF- ⁇ when incubated with T2 cells preincubated with M9-2 or M10-3 (data not shown). Therefore, M9-2 or M10-3 are common epitopes recognized by a majority of HLA-A2 restricted melanoma specific TIL.
  • MART-1 is a 118 amino acid protein containing a single transmembrane domain and is expressed in most melanoma cells as well as cultured melanocytes and retina similar: to the expression pattern of the cDNA for a form of gp100 described in Example 3.
  • the gp100 is recognized by 4 of 10 TIL.
  • peptide M9-2 most effectively sensitized T2 cells for lysis ( FIG. 2 ) suggesting that this peptide may be naturally processed and presented on tumor cells.
  • the T-cells recognizing M9-2 may react with peptide M10-3 or M10-4 because the latter 10-mer peptides contain the 9 amino acid sequence of peptide M9-2.
  • M10-4 was poorly recognized by the T-cell clone A42, but was well recognized by some TIL lines, although a higher concentration of M10-4 was necessary to observe the lysis.
  • TIL lines may contain different T-cell clones which only recognize either M9-2 or M10-4.
  • Peptides M10-3 and M10-4 may also be naturally processed and presented by tumor cells.
  • the existence of multiple melanoma antigens presented by HLA-A2 has previously been suggested by analyzing the recognition of melanoma cell clones by a variety of T-cell clones (Knuth, A. et al. (1989), Proc. Natl. Acad. Sci. (USA) 86:2804, Wolfel, T. et al., 1989 J. Exp. Med.
  • the cDNA25 clone encoding a form of the melanoma antigen designated gp100 was cloned by techniques similar to those described in Example 1 and in Miki, T., et al. (1991) Proc. Natl. Acad. Sci. (USA) 88:5167-5171. Briefly, a breast cancer cell line, MDA231 (ATCC #HTB26), transfected with a cDNA library in ⁇ pCEV27 made from the 501mel melanoma cell line was screened for antigen positivity by measuring interferon- ⁇ (IFN- ⁇ ) secretion when cocultured with TIL1200. TIL1200 was generated as described in Kawakami, Y., (1988), J.
  • IFN- ⁇ interferon- ⁇
  • the integrated cDNA was recovered from the genomic DNA of positive transfectants by PCR and cloned into the mammalian expression plasmid pcDNA3 (Invitrogen, San Diego, Calif.).
  • the full length cDNA for cDNA25 was isolated from the 501mel ⁇ pCEV27 library using the cDNA25 probe.
  • the ⁇ phage containing the full length cDNA25 was digested with XhoI, and then self-ligated with T4 DNA ligase to make the plasmid pCEV27-FL25.
  • a full length cDNA25 isolated by PCR using the specific primers designed for gp100 was cloned in pCRII(Invitrogen), and then cloned into pcDNA3(pcDNA3-FL25).
  • pcDNA3 pcDNA3-FL25
  • DNA sequences of the plasmid clones were determined with an automated DNA sequencer (Model 373A; Applied Biosystems, Inc.), using the Taq DyeDeoxy terminator cycle sequencing kit (Applied Biosystems, Inc.) using the manufacturer's instructions.
  • Peptides were synthesized by a solid phase method using a Gilson AMS 422 multiple peptide synthesizer.
  • the peptides were purified by HPLC on a Vydac C-4 column with 0.05% TFA/water-acetonitrile.
  • TIL lysis of T2 RET-cells preincubated with peptides for 2 hour (h) was measured using a 51 Cr release cytotoxicity assay.
  • a 29 year old male patient, designated patient number 1200, with a widely metastatic melanoma who had previously failed chemotherapy and radiation therapy was treated with a single preparatory dose of 25 mg/Kg cyclophosphamide followed by the intravenous infusion of 1.6 ⁇ 10 11 TIL (including 9.1 ⁇ 10 9 Indium-ill labeled TIL) plus 7 doses of IL-2 at 720,000 IU/Kg given every 8 hours.
  • TIL including 9.1 ⁇ 10 9 Indium-ill labeled TIL
  • 7 doses of IL-2 at 720,000 IU/Kg given every 8 hours.
  • a second cycle of treatment with TIL and IL-2 was given three weeks later. Radionuclide scans showed localization of TIL in rumor deposits ( FIG. 3A ).
  • Biopsy of subcutaneous tumors on days 8 and 11 after treatment showed significant localization of TIL to tumor (ratios of injectate per gram in tumor compared to normal tissue were 14.9 and 14.0 respectively).
  • the patient's cancer regressed rapidly following the first course of treatment.
  • 2 of 3 liver lesions had disappeared and a third lesion shrank by 50%.
  • Multiple subcutaneous metastases regressed completely as shown in FIG. 3B (the product of perpendicular diameters of individual lesions are shown).
  • TIL1200 Characterization of in vitro function of TIL1200
  • TNF ⁇ tumor necrosis factor-alpha
  • GM-CSF granulocyte-macrophage colony stimulating factor
  • a CD8 + CTL line, TIL1200 established from a metastatic subcutaneous tumor mass of patient 1200, lysed fresh autologous melanoma cells as well as 10 of 15 HLA-A2 + allogeneic melanoma cell lines, but did not lyse 16 of 18 HLA-A2 ⁇ melanoma cell lines or 6 of 8 HLA-A2 + non-melanoma cell lines (Shilyansky, J., et al. (1993) Proc. Natl. Acad. Sci. USA, 91, 2829-2833, unpublished data).
  • Table 7 shows a cytotoxicity assay against 5 representative HLA-A2 + melanoma cell lines that were lysed by TIL1200, 4 representative HLA-A2+ melanoma cell lines that were not lysed by TIL1200, and one HLA-A2 ⁇ melanoma cell line.
  • TIL1200 also secreted IFN- ⁇ when cocultured with HLA-A2 + normal cultured melanocytes established from neonatal foreskin as well as HLA-A2 + melanoma cell lines (Table 8). Therefore, TIL1200 appeared to recognize a non-mutated self peptide expressed in most melanomas and cultured neonatal melanocytes in an HLA-A2 restricted fashion.
  • G418 resistant cells were selected and approximately 6700 individual transfectants from each cell line were isolated and screened based on their ability to stimulate IFN- ⁇ secretion from TIL1200.
  • TIL1200 Lysis of HLA-A2 + gp100 + Melanoma Cell Lines TIL1200 LAK gp100 (% specific Target HLA-A2 FACS Northern lysis) 501mel + + 46 78 526mel + + + 39 74 624mel + + + 33 76 952mel + + + 25 76 Malme3M + + + 43 70 C32 + ⁇ ⁇ /+* 6 82 RPMI7951 + ⁇ ⁇ 9 67 WM115 + ⁇ ⁇ 5 68 HS695T + ⁇ ⁇ 3 87 397mel ⁇ + + 0 70 5 hour (h) 51 Cr release assay was performed to measure cellular cytotoxicity at an effector: target ratio of 40:1 as previously described(Kawakami, Y.
  • HLA-A2 and gp100 recognized by monoclonal antibody HMB45 was measured by flow cytometry(FACS).
  • the expression of gp100 RNA was analyzed by Northern blot with a cDNA25 probe. * ⁇ /+ indicates very weak positive.
  • TIL1200 Recognition of HLA-a-a2 + Neonatal Melanocytes TIL1200 TIL888 Stimulator HLA-A2 (pgIFN- ⁇ /ml) 501mel + 562 0 624mel + 439 0 397mel ⁇ 0 0 888mel ⁇ 0 1970 NHEM493 ⁇ 441 0 NHEM527 + 418 0 NHEM530 + 164 0 NHEM616 + 53 0 FM725 + 107 0 FM801 + 250 343 NHEM483 ⁇ 0 0 NHEM680 ⁇ 0 0 HA002 ⁇ 0 0 0 The IFN- ⁇ secretion by TIL was measured by ELISA as previously described in Example 1.
  • TIL888 is a class I MHC restricted melanoma specific CTL, not restricted by HLA-A2.
  • NHEM, FM, and HA refer to normal cultured melanocyte cell lines, all others are melanoma cell lines.
  • HLA-A2 expression was determined by flow cytometry and interferon-gamma secretion was measured by ELISA.
  • 3 cDN25 clones that were independently amplified by PCR revealed that cDNA25 was distinct from two previously registered genes, a gp100 (GenBank Access No. M77348) and Pmel17 (Kwon, B. S., et al. (1991) Proc. Natl. Acad.
  • the cDNA 25 differed from the gp100 in GenBank (Accession No. M77348, also known as gp95) by two nucleotides, from the PMEL 17 sequence (Kwon et al. (1991) Proc. Natl. Acad. Sciences (USA) 58: 9228-9232) by 3 bases and a 21 base pair deletion. ( FIG. 5B ).
  • the full length cDNA25(FL25) was isolated in two plasmids, pCEV27-FL25 or pcDNA3-FL25.
  • Transfection of either plasmid into COS7 along with pcDNA3-HLA-A2.1 conferred to COS7 the ability to induce IFN- ⁇ secretion by TIL1200.
  • the amount of IFN- ⁇ secretion stimulated by COS7 transfected with the full length DNA plus HLA-A2.1 was similar to that stimulated by 501mel and was higher than that stimulated by COS7 transfected with the truncated cDNA25 possibly due to improved translation starting at the normal AUG initiation codon (Table 9, Exp.2 and 3).
  • the 5′ region missing from the truncated cDNA25 may contain other epitopes recognized by clones in TIL1200.
  • the nucleotide and corresponding amino acid sequences of the truncated cDNA 25 and the full length cDNA25 cloned from the 501mel cDNA library by screening with the cDNA25 probe were compared with the GenBank sequences of Pmel17 isolated from normal melanocytes and gp100 isolated from the melanoma cell line MEL-1. ( FIG. 5B ).
  • the full length cDNA25 differed from the gp100 amino acid sequence at position 162. This amino acid difference is possibly caused by polymorphism or mutation in the tumor.
  • cDNA25 had 2 amino acid differences at positions 162 and 274, compared to Pmel17 and did not contain 7 amino acids that existed in Pmel17 at positions 588-594.
  • the amino acid sequence of the truncated cDNA25 that was isolated from the original MDA231 transfectant has a different sequence at the 3′ end (from position 649 to the end) due to a frame shift caused by one extra cytidylic acid. It is not clear whether this difference was due to a true allelic difference or to a mutation that occurred during manipulation of the DNA. Nevertheless, TIL1200 appeared to recognize non-mutated peptides located between position 236 and 648.
  • cDNA25 also had 87% similarity in amino acid sequence to cDNA RPE1 (Kim, R., and Wistow, G. J. (1992) Exp. Eve Res.
  • a gp100 protein was known to be recognized by monoclonal antibody HMB45 (Adema et al., (1993) Am. J. Patholoay, 143: 1579-1585).
  • COS7 cells transfected with the full length cDNA25 were evaluated by flow cytometry using this monoclonal antibody. After transient expression of either pCEV27-FL25 or pcDNA3-FL25, COS7 expressed the antigen detected by HMB45 (data not shown).
  • T-cell T-cell
  • B-cells 501EBVB, 836EBVB
  • fibroblast M1
  • 20 non-melanoma tumor cell lines colon cancer, Collo, SW480, WiDr; breast cancer, MDA231, MCF7, HS578, ZR75; neuroblastoma, SK—N-AS, SK—N—SH; Ewing sarcoma, TC75, RD-ES, 6647; sarcoma 143B; glioma, U138MG, HS683; renal cell cancer, UOK108, UOK117, small cell lung cancer, H1092; Burkitt's lymphoma, Daudi; myeloma HMY) were all negative for cDNA25 (data not shown).
  • this gene appeared to be specifically expressed in melanocyte lineage cells, consistent with the expression pattern of previously isolated forms of gp100 when analyzed using monoclonal antibodies, HMB45, NKI/betab, or HMB-50 (Adema, G. J., et al. (1993) Am J Pathology 143: 1579-1585; Gown, A. M., et al., ( 1986 ) Am J Pathol 123:195-203; Colombari, R., et al. (1988) Virchows Archiv A Pathol Anat. 413:17-24); Vennegoor, C., et al. (1988) Am. J. Pathol. 130:179-192; Vogel, A. M., and Esclamado R.
  • TIL TIL 1200
  • TIL TIL1200
  • TIL1200 which was a TIL line containing multiple CTL species recognized a tumor antigen in the context of HLA-A2, which is the most frequently expressed class I MHC antigen (about 50% of individuals) and has been shown to be a dominant restriction element for the induction of melanoma specific CTL. (Crowley, N. J., et al. (1991) J. Immunol. 146, 1692-1699).
  • a cDNA ( FIG. 4 ; SEQ ID NO: 26) encoding an antigen recognized by TIL1200 and identified as a form of gp100, a membrane glycoprotein also recognized by monoclonal antibodies, BMB45, HMB50 or NKI/betab has been identified.
  • a cDNA ( FIG. 4 ; SEQ ID NO: 26) encoding an antigen recognized by TIL1200 and identified as a form of gp100, a membrane glycoprotein also recognized by monoclonal antibodies, BMB45, HMB50 or NKI/betab has been identified.
  • a cDNA encoding another form of gp100 was also isolated by screening with a rabbit polyclonal antiserum against gp100 (Adema, G. J., et al. (1993) Am J Pathology 143:1579-1585) and TIL1200 also lysed HLA-A2 + cell lines transfected with this cDNA clone (Bakker, A. B. H. et al. (1994) J. Exp. Med. 179:1005-1009).
  • T cells reactive to the self-antigen gp100 in tumors and the possible enrichment of these T cells at the tumor site as a possible consequence of the specific accumulation and expansion of antigen reactive cells raises important questions about the nature of the immune response to self antigens on growing cancers and about the mechanisms of immunologic tolerance to self-antigens.
  • the gp100 protein ( FIG. 5A ; SEQ ID NO: 27) and the ten amino acid peptide identified may represent a human tumor rejection antigen since the transfer into patient 1200 of TIL1200 plus IL2 was associated with cancer regression.
  • the traffic of TIL1200 to tumor deposits in vivo and the rapidity of the antitumor response are characteristics of the response to TIL therapy, although IL2 may also have been involved in the tumor rejection.
  • Adoptive transfer of 3 other TIL lines which recognized gp100 as well as MART-1 also mediated tumor regression (data not shown).
  • Tyrosinase (Brichard, V., et al. (1993) J. Exp. Med. 178, 489-495) and MART-1 (see Example 1) have been identified as melanoma antigens recognized by HLA-A2 restricted CTL.
  • Another antigen, MAGE-1 is recognized by HLA-A1 restricted melanoma-specific CTL and is expressed on a variety of cancer cells as well as testis (Van Der Bruggen, P. et al. (1991) Science, 254:1643-1647).
  • HLA-A2 restricted TIL none of the ten HLA-A2 restricted TIL recently developed appeared to recognize MAGE-1 (Zakut, R., et al. (1993) Cancer Res. 53: 5-8).
  • gp100 proteins in melanomas the recognition of a peptide by T cells infiltrating into tumor, its restriction by HLA-A2, present in 50% of individuals, and the association of anti gp100 reactivity with cancer regression in patient 1200 imply that the gp100 antigen in particular the novel immunogenic peptides derived from the gp100 amino acid sequence ( FIG. 5A ; SEQ ID NO: 27) may be particularly useful for the development of active immunotherapies for patients with melanoma.
  • Peptides were synthesized by a solid phase method using a peptide synthesizer (model AMS 422;Gilson Co.Inc., Worthington, Ohio)(>90% purity).
  • the peptides to be synthesized were selected from the reported human sequence of gp100 based on HLA-A2.1 binding motifs (Falk, K., (1991) Nature 351:290; Hunt, D. F., et al, (1992) Science 255:1261; Ruppert, J., et al., (1993) Cell 74:929; Kubo, R T, et al. (1994) J Immunol. 152:3913).
  • Soluble HLA-A2.1 heavy chain, human beta 2-microglobulin, radiolabeled peptide HBc 18-27 (FLPSDYFPSV) and various concentrations of the sample peptides were coincubated in the presence of protease inhibitors for 2 days at room temperature as previously described (Ruppert, J., et al., (1993) Cell 74:929; Kubo, R T, et al., (1994). J Immunol. 152:3913; Sett A., et al., (1994). Molecular Immunol. 31:813).
  • the percentage of labeled peptide bound to HLA-A2.1 was calculated after separation by gel filtration and the concentration of the sample peptide necessary to inhibit 50% of the binding of the labeled peptide was calculated.
  • the relative affinity of peptides to HLA-A2.1 were also calculated as a ratio (concentration of the standard HBc 18-27 peptide to inhibit 50% of the binding of the labeled peptide/concentration of the sample peptide to inhibit 50% of the binding of the labeled peptide) as previously described (Sett A., et al., (1994) Molecular Immunol. 31:813).
  • Peptide binding was defined as high (50% inhibition at ⁇ 50 nM, ratio>0.1), intermediate (50-500 nM, ratio 0.1-0.01) or weak (>500 nM, ratio ⁇ 0.01) (Ruppert, J., et al., (1993) Cell 74:929; Kubo, R T, et al., (1994) J Immunol. 152:3913; Sett A., et al., (1994) Molecular Immunol. 31:813).
  • the pcDNA3 plasmid containing the full length gp100 cDNA ( FIG. 4 , Example 3; Kawakami, Y., et al., (1994)). Proc Natl Acad Sci (USA) 91:6458) was digested with Xho I and Xba I. After incorporation of alpha-phosphorothioate deoxynucleoside triphosphate into the Xba I site, a standard exonuclease III nested deletion was performed using the Exo Size Deletion Kit (New England Biolabs, inc., Beverly, Mass.). The deleted clones were self-ligated and amplified. The exact deletion for each clone was confirmed by DNA sequencing.
  • pcDNA3 plasmids (Invitrogen, San Diego Calif.) containing the cDNA fragments (D3, D5, D4, C3) generated by the sequential deletion with exonuclease from the 3′ end of the full length gp100 cDNA as well as the truncated gp100 cDNA lacking the 5′-coding region (25TR) (Example 63; Kawakami, Y.
  • a 51 Cr release assay or an IFN- ⁇ release assay were performed as previously described (Examples 1 and 2; Kawakami, Y., et al., (1994)). Proc Natl Acad Sci (USA) 91:3515; Kawakami, Y., et al., (1988). J. Exp. Med. 168:2183). Either COS7 cells transfected with cDNA encoding melanoma antigens and HLA-A2.1 cDNA, or T2 cells preincubated with peptides were used as stimulators for the IFN- ⁇ release assay. T2 cells pulsed with peptides were also used as targets for cytotoxicity assays (Kawakami, Y., (1994) J. Exp. Med. 180:347).
  • TIL620 (620-1, 620-2) or TIL660 (660-1, 660-2, 660-3) were grown from the TIL culture that was administered into the autologous patient, but they were separately cultured and had slightly different specificities likely due to the in vitro expansion of different clones.
  • G9 209 (ITDQVPFSV) (SEQ ID NO:48)
  • G10 209 (TITDQVPFSV) (SEQ ID NO:49)
  • G9 154 (KTWGQYWQV) (SEQ ID NO:46) and G10 154 (KTWGQYWQVL) (SEQ ID NO:47), which has an extra leucine at the C-terminus of G9 154 , were recognized by TIL1200, TIL620-2 and TIL660-2.
  • G10-4 (LLDGTATLRL) (SEQ ID NO:33) was recognized by TIL1200 as demonstrated (Example 3).
  • the peptide G9 280 (YLEPGPVTA) (SEQ ID NO:40) was recognized by TIL660 and TIL1143.
  • TIL660-3 also recognized G10-5 (VLYRYGSFSV) (SEQ ID NO:34) as well as G9280. Lysis of T2 cells preincubated with G10-5 was repeatedly low, possibly because a small subset of T-cell clones was specific for this epitope.
  • TIL 1200 recognized COS cells transfected with the fragments, 25TR, D5, D4 or C4, but not with D3, suggesting that at least 2 epitopes existed in the regions of amino acid residues 146-163 and 236-661.
  • G9 154 and G10 154 were the only peptides which contained HLA-A2.1 binding motifs in the region 146-163 and both were recognized by TIL1200.
  • G10-4 was located in the region 236-661 and was recognized by TIL1200.
  • TIL620-1 recognized COS cells transfected with C4 but not with D3, D5, D4 or 25TR, suggesting that the epitope existed within residues 187-270.
  • TIL620-1 Another subculture of TIL620, also recognized COS cells transfected with D5 and D4, but not D3, and recognized G9 154 and G20 154 in the region 147-163, also recognized by TIL1200.
  • TIL660-1 and TIL1143 recognized COS cells transfected with C4 or 25TR, but not with D3, D5, or D4, suggesting that epitopes existed in the 2 regions 187-270 and 236-661.
  • G10-5 appeared to be inhibitory to the cytotoxic activity of CTL at concentration greater than 10 ng/ml since lysis of T2 cells incubated with G10-5 at more than 10 ng/ml was repeatedly lower than at 1-10 ng/ml in this assay condition in which the peptide was present in the medium during entire 4 h cytotoxicity assay ( FIG. 8D ).
  • the relative binding affinity of these epitopes to HLA-A2.1 was also measured using an in vitro competitive binding assay (Table 13).
  • G9 154 had an higher binding affinity (50% inhibition of the standard peptide at 11 nM) to the HLA-A2.1 molecule than G10 154 (1010 nM) which contains an extra leucine at the C-terminus of G9 154 , and could sensitize T2 cells at lower concentrations than G10 154 ( FIG. 8A ).
  • G9 209 also bound to HLA-A2.1 with higher affinity (84 nM) than G10 208 (2080 nM) which contains an extra threonine at the N-terminus, and could sensitize T2 cells at lower concentrations of peptide than G10 208 ( FIG. 8B ).
  • the 9-mer peptides were superior to the corresponding 10 mer peptides in the sensitization of T2 cells to CTL lysis, and they also had higher binding affinities to HLA-A2.1. This was also the case for the identified MART-1 9 and 10 amino acid peptides (M9-2, M10-3, M10-4) (Example 2; Kawakami, Y., et al., (1994). J. Exp. Med. 180:347).
  • the results of the peptide titration in the T2 cell lysis assay correlated with the results of the HLA-A2.1 binding affinity as measured by the in vitro binding assay.
  • the other gp100 epitopes, G9 280 , G10-4, or G10-5 had binding affinities for HLA-A2.1 with 50 inhibition at 95 nM, 483 nM, or 13 nM, respectively.
  • the HLA-A2.1 binding affinities of the previously identified HLA-A2 restricted melanoma epitopes in MART-1 (Example 2; Kawakami, Y., et al., (1994) J. Exp. Med. 180:347) and tyrosinase (Wolfel, T., (1994) Eur. J. Immunol.
  • the MART-1 peptide M9-2 may also be designated M9 27
  • the MART-1 peptide M10-3 may also be designated M10 26
  • the MART-1 peptide M10-4 may also be designated M10 27
  • the gp100 peptide G10-4 may also be designated G10 457 and the gp100 peptide G10-5 may also be designated G10 476 .
  • TIL620-1, -2 or TIL660-1, -2, -3 were grown from the same TIL which was administered into the autologous patient, but were separately cultured. 624mel, HLA-A2+ gp100+, MART-1+ melanoma cell line, 397mel, HLA-A2 ⁇ melanoma cell line. T2 cells, HLA-A2+ T cell-B-cell hybridoma. Bold: statistically significant lysis
  • Peptide synthesis and HLA-A2.1 binding assay Peptides were synthesized by a solid phase method using a multiple peptide synthesizer and purified by HPLC, as previously described (Rivoltini, L et al. (1995) Journal of Immunology Volume 154:2257-2265).
  • the relative binding of peptides to HLA-A2.1 based on the inhibition of binding of a radiolabeled standard peptide to detergent-solubilized MHC molecules, was performed as previously described (Rivoltini, L et al. (1995) Journal of Immunology Volume 154:2257-2265).
  • test peptides ranging from 100 ⁇ M to 1 nM
  • 5 nM radiolabeled Hbc 18-27 (FLPSDYFPSV) peptide and HLA-A2.1 heavy chain and ⁇ 2-microglobulin were coincubated together with the 5 nM radiolabeled Hbc 18-27 (FLPSDYFPSV) peptide and HLA-A2.1 heavy chain and ⁇ 2-microglobulin for 2 days at room temperature in the presence of protease inhibitors.
  • the percentage of MHC-bound radioactivity was determined by gel filtration and the 50% inhibitory dose was calculated for each peptide.
  • PBMC peripheral blood of HLA-A2 + melanoma patients and normal donors by centrifugation on Ficoll-Hypaque gradients and used as fresh or cryopreserved samples.
  • Peptide specific CTL lines were generated as follows: at day 0, PBMC were plated at a concentration of 1.5 ⁇ 10 6 /ml in 24-well plates (2 ml/well) in Iscove's medium containing 10% human AB serum, L-glutamine, antibiotics (CM) and in the presence of 1 ug/ml peptide. Two days later, 12 IU/ml interleukin 2 (IL-2) (Chiron Co., Emeryville, Calif.) were added to the cultures.
  • IL-2 interleukin 2
  • Lymphocytes were then restimulated weekly as follows: responder cells were harvested, washed once and replated in 24-well plates at a concentration of 2.5 ⁇ 10 5 cells/ml in CM. Autologous PBMC were thawed, washed twice in PBS, resuspended at 5-8 ⁇ 10 6 cells/ml in CM and pulsed with 1 ug/ml peptide in 15-ml conical tubes (5 ml/tube) for 3 hours at 37° C. These PBMC (stimulators) were then irradiated at 3000 rads, washed once in PBS and added to the responder cells at responder:stimulator ratios ranging between 1:3 and 1:10.
  • IL-2 12 IU/ml IL-2 were added to the cultures.
  • the activity of these CTL was tested by cytotoxicity assays after at least 2 rounds (14 days) of peptide stimulation.
  • the dissociated tumor suspension were cultured for 1-2 days in 10% FCS RPMI-1640 medium to allow tumor cell adherence.
  • the lymphocytes, recovered from the non-adherent fraction, were used for the induction of peptide specific CTL as described above.
  • 51 Cr release cytotoxic assays were performed to detect the recognition of peptide and melanoma cells by CTL.
  • T2 cell lines were preincubated for 2 h at 37° C. with 1 ug/ml peptide, washed and used as target cells in a 51 Cr release cytotoxic assay.
  • the melanoma lines 624mel was established in our laboratory (See Example 1).
  • the isolated MART-1 or gp100 melanoma epitopes contain non-dominant amino acid at major anchor position such as alanine (the 2nd position of M9-2, the 9th position of G9-280) and threonine (the 2nd position of G9-154 and G9-209).
  • the M9-2, G9-209 and G9-280 are not high affinity binders.
  • PBL stimulated with autologous PBMC pulsed with G9-154-2I, G9-209-1F2L, or G9-280-9V (Tables 18, 19 and 20) recognized and lysed not only the original epitopes but also melanoma tumor cells (624mel) better than PBL stimulated with natural epitopes (G9-154, G9-209, G9-280).
  • modified peptides could be used for induction of anti-tumor T-cells instead of natural epitopes.
  • Other peptides which were not recognized by the particular T-cells used in our study, but have higher binding affinity to HLA-A2.1 may induce a different set of T-cells capable of recognizing the original melanoma epitopes in in vitro or in vivo.
  • These modified peptides may be used for induction of anti-melanoma T-cells in vitro and immunization of patients for the treatment of patients with melanoma or for the prevention of melanoma.
  • MART-1 vaccines may be efficacious in treating mammals afflicted with melanoma.
  • MART-1 vaccines may be administered to individuals.
  • Mammals can be immunized with the MART-1 proteins, peptides or modified peptides described herein in ranges of about 1 mg-to to about 100 mg.
  • mammals, preferably humans may be immunized with the MART-1 nucleic acid sequence inserted into a viral vector such as vaccinia virus, adenovirus or fowl pox virus.
  • a range of about 10 6 to about 10 11 viral particles carrying the MART-1 nucleic acid sequences corresponding to immunogenic MART-1 peptides or modified peptides or analogs therof, may be administered per mammal, preferably a human.
  • the mammals will be monitored for antibodies to the immunogen or increase in cytotoxic lymphocytes (CTL) recognizing the immunogen by conventional methods or alleviation of clinical signs and symptoms of the active disease.
  • CTL cytotoxic lymphocytes
  • Specific parameters to be assessed include production of immune cells that recognize the vaccine antigen or tumor regression.
  • Such vaccines may be administered either prophylactically or therapeutically.
  • Mammals may also be immunized with the gp-100 nucleic acid sequence inserted into a retroviral vector or GP-100 immunogenic peptides or modified peptides or analogs thereof. Suggested dose ranges of the antigen in retroviruses that may be used are about 10 6 to about 10 11 viral particles per mammal, preferably a human. Response and efficacy of the retroviral vaccines will be assessed as described above.
  • Lymphocytes Sensitized to Immunogenic Peptides Derived from Melanoma Antigens for Therapeutically Treating Mammals Afflicted with Melanoma
  • T-lymphocytes presensitized to the melanoma antigen may be effective in therapeutically treating mammals afflicted with melanoma.
  • the T-lymphocytes will be isolated from peripheral blood lymphocytes or tumor infiltrating lymphocytes and exposed in vitro to the MART-1 protein or peptide.
  • T-lymphocytes are isolated from peripheral blood or melanoma tumor suspensions and cultured in vitro (Kawakami, Y. et al. (1988) J. Exp. Med. 168: 2183-2191).
  • the T-lymphocytes are exposed to the MART-1 peptide AAGIGILTV for a period of about to 1-16 hours at a concentration of about 1 to about 10 mg/ml.
  • T-lymphocytes exposed to the antigen will be administered to the mammal, preferably a human at about 10 9 to about 10 12 lymphocytes per mammal.
  • the T-lymphocytes may be exposed to the modified MART-1 peptides.
  • the lymphocytes may be administered either intravenously, intraperitoneally or intralesionally. This treatment may be administered concurrently with other therapeutic treatments such as cytokines, radiotherapy, surgical excision of melanoma lesions and chemotherapeutic drugs, adoptive T lymphocyte therapy.
  • the T-lymphocytes may be exposed to the gp100 immunogenic peptides or modified immunogenic peptides described herein.

Abstract

The present invention provides a nucleic acid sequence encoding a melanoma antigen recognized by T lymphocytes, designated MART-1. This invention further relates to bioassays using the nucleic acid sequence, protein or antibodies of this invention to diagnose, assess or prognoses a mammal afflicted with melanoma or metastata melanoma. This invention also provides immunogenic peptides derived from the MRT-1 melanoma antigen and a second melanoma antigen designated gp100. This invention further provides immunogenic peptides derived from the MART-1 melanoma antigen or gp100 antigen which have been modified to enhance their immunogenicity. The proteins and peptides provided can serve as an immunogen or vaccine to prevent or treat melanoma.

Description

  • This application is a divisional of application Ser. No. 09/898,860, filed Jul. 3, 2001, which is a divisional of application Ser. No. 09/267,439, filed Mar. 12, 1999, now U.S. Pat. No. 6,270,778, which is a divisional of application Ser. No. 09/073,138, filed May 5, 1998, now U.S. Pat. No. 6,537,560, which is a continuation-in-part of application Ser. No. 08/417,174, filed Apr. 5, 1995, now U.S. Pat. No. 5,844,075, which is a continuation-in-part of application Ser. No. 08/231,565, filed Apr. 22, 1994, now U.S. Pat. No. 5,874,560, all of which are hereby incorporated by reference.
  • FIELD OF THE INVENTION
  • This invention is in the field of prevention and treatment of human cancers. More specifically, this invention relates to genes encoding melanoma antigens recognized by T-Cells and their corresponding proteins and to preventative, diagnostic and therapeutic applications which employ these genes or proteins.
  • BACKGROUND OF THE INVENTION
  • Melanomas are aggressive, frequently metastatic tumors derived from either melanocytes or melanocyte related nevus cells (“Cellular and Molecular Immunology” (1991) (eds) Abbas A. K., Lechtiman, A. H., Pober, J. S.; W. B. Saunders Company, Philadelphia: pages 340-341). Melanomas make up approximately three percent of all skin cancers and the worldwide increase in melanoma is unsurpassed by any other neoplasm with the exception of lung cancer in women (“Cellular and Molecular Immunology” (1991) (eds) Abbas, A. K., Lechtiman, A. H., Pober, J. S.; W.B. Saunders Company Philadelphia pages: 340-342; Kirkwood and Agarwala (1993) Principles and Practice of Oncology 7:1-16). Even when melanoma is apparently localized to the skin, up to 30% of the patients will develop systemic metastasis and the majority will die (Kirkwood and Agarwala (1993) Principles and Practice of Oncology 7:1-16). Classic modalities of treating melanoma include surgery, radiation and chemotherapy. In the past decade immunotherapy and gene therapy have emerged as new and promising methods for treating melanoma.
  • T cells play an important role in tumor regression in most murine tumor models. Tumor infiltrating lymphocytes (TIL) that recognize unique cancer antigens can be isolated from many murine tumors. The adoptive transfer of these TIL plus interleukin-2 can mediate the regression of established lung and liver metastases (Rosenberg, S. A., et al., (1986) Science 233:1318-1321). In addition, the secretion of IFN-γ by injected TIL significantly correlates with in vivo regression of murine tumors suggesting activation of T-cells by the tumor antigens. (Barth, R. J., et al., (1991) J. Exp. Med. 173:647-658). The known ability of tumor TIL to mediate the regression of metastatic cancer in 35 to 40% of melanoma patients when adoptively transferred into patients with metastatic melanoma attests to the clinical importance of the antigens recognized (Rosenberg, S. A., et al., (1988) N Engl J Med 319:1676-1680; Rosenberg S. A. (1992) J. Clin. Oncol. 10:180-199).
  • T cell receptors on CD830 T cells recognize a complex consisting of an antigenic peptide (9-10 amino acids for HLA-A2), β-2 microglobulin and class I major histocompatibility complex (MHC) heavy chain (HLA-A, B, C, in humans). Peptides generated by digestion of endogenously synthesized proteins are transported into the endoplastic reticulum, bound to class I MHC heavy chain and β2 microglobulin, and finally expressed in the cell surface in the groove of the class I MHC molecule. Therefore, T cells can detect molecules that originate from proteins inside cells, in contrast to antibodies that detect intact molecules expressed on the cell surface. Therefore, antigens recognized by T cells may be more useful than antigens recognized by antibodies.
  • Strong evidence that an immune response to cancer exists in human—is provided by the existence of lymphocytes within melanoma deposits. These lymphocytes, when isolated, are capable of recognizing specific tumor antigens on autologous and allogeneic melanomas in an MHC restricted fashion. (Itoh, K. et al. (1986), Cancer Res. 46: 3011-3017; Muul, L. M., et al. (1987), J. Immunol. 138:989-995); Topalian, S. L., et al., (1989) J. Immunol. 142: 3714-3725; Darrow, T. L., et al., (1989) J. Immunol. 142: 3329-3335; Hom, S. S., et al., (1991) J. Immunother. 10:153-164; Kawakami, Y., et al., (1992) J. Immunol. 148: 638-643; Hom, S. S., et al., (1993) J. Immunother. 13:18-30; O'Neil, B. H., et al., (1993) J. Immunol. 151: 1410-1418). TIL from patients with metastatic melanoma recognize shared antigens including melanocyte-melanoma lineage specific tissue antigens in vitro (Kawakami, Y., et al., (1993) J. Immunother. 14: 88-93; Anichini, A. et al., (1993) et al., J. Exp. Med. 177: 989-998). Anti-melanoma T cells appear to be enriched in TIL probably as a consequence of clonal expansion and accumulation at the tumor site in vivo (Sensi, M., et al., (1993) J. Exp. Med. 178:1231-1246). The fact that many melanoma patients mount cellular and humoral responses against these tumors and that melanomas express both MHC antigens and tumor associated antigens (TAA) suggests that identification and characterization of additional melanoma antigens will be important for immunotherapy of patients with melanoma.
  • Peripheral blood lymphocytes have been used to identify potential melanoma tumor antigens. Van Der Bruggen et al. (1991) Science 254: 1643-1647 has characterized a gene coding for a melanoma antigen, designated MAGE-1, using T cell clones established from the peripheral blood of patients who were repetitively immunized in vivo with mutagenized tumor cells. Cytotoxic T-cells derived from the peripheral blood lymphocytes of patients with melanoma were used to identify a potential antigenic peptide encoding MAGE-1 (Traversari, C., et al. (1992) J. Exp. Med. 176:1453-1457). Brichard et al. (1993) J. Exp. Med. 178:489-495 has also characterized a gene encoding a melanoma antigen designated tyrosinase using peripheral blood lymphocytes from patients who were sensitized by repetitive in vitro stimulation with tumor. Further support for the therapeutic potential of melanoma antigens is provided by Brown et al. (U.S. Pat. No. 5,262,177). Brown et al. (U.S. Pat. No. 5,262,177) relates to a recombinant vaccinia virus-based melanoma vaccine where the melanoma antigen p97 is reported to show a protective effect from tumor cell challenge in a murine model. Characterization of additional melanoma antigens is important for the development of new strategies for cancer immunotherapy, in particular for melanoma.
  • SUMMARY OF THE INVENTION
  • This invention relates, in general, to nucleic acid sequences encoding melanoma antigens recognized by T-lymphocytes (MART-1) and protein and peptides encoded by these sequences. This invention further provides bioassays for these nucleic acid sequences, proteins and peptides. This invention also provides peptides which have been derived from the MART-1 amino acid sequence and modified to enhance their immunogenocity. This invention also provides therapeutic uses for the nucleic acid sequences, proteins, peptides or modified peptides described herein.
  • It is a general object of the present invention to provide a substantially purified and isolated nucleic acid sequence which encodes for the MART-1 melanoma antigen.
  • It is another object of this invention to provide a recombinant molecule comprising a vector and all or part of the nucleic acid sequence encoding MART-1.
  • It is another object of this invention to produce recombinant proteins encoded by all or part of the nucleic acid sequence encoding MART-1.
  • It is a further object of this invention to provide monoclonal or polyclonal antibodies reactive with the MART-1 protein, peptides or portions thereof.
  • It is an object of this invention to provide methods of detecting the MART-1 gene or MART-1 mRNA in a biological sample.
  • It is another object of this invention to provide methods of detecting the MART-1 protein or peptides in a biological sample.
  • It is an object of this invention to provide diagnostic methods for human disease, in particular for melanomas and metastatic melanomas.
  • It is a further object of this invention to provide methods for prophylactic or therapeutic uses involving all or part of the nucleic acid sequence encoding MART-1 and its corresponding protein or peptides derived from the MART-1 amino acid sequence.
  • It is also an object of this invention to provide melanoma vaccines comprising all or part of the nucleic acid sequence encoding MART-1 or its corresponding protein for preventing or treating melanoma.
  • It is a further object of this invention to provide immunogenic peptides derived from the MART-1 protein sequence for use in vaccines.
  • It is yet another object of this invention to provide peptides derived from the MART-1 protein sequence which have been modified to increase their immunogenicity or enhance induction of antimelanoma immune response by enhancing their binding to MHC molecules, for use in the prophylactic or therapuetic mothods described herein.
  • In addition, it is another object of this invention to provide multivalent vaccines comprising all or part of the MART-1 nucleic acid sequence or its corresponding protein or peptides and at least one other immunogenic molecule capable of eliciting the production of antibodies in a mammal to melanoma antigens.
  • It is another object of this invention to provide a method for preventing or treating melanoma utilizing all or part of the MART-1 nucleic acid sequence or its corresponding protein in gene therapy protocols.
  • It is a further object of this invention to provide immunogenic peptides derived from a gp100 melanoma antigen protein sequence for use in vaccines.
  • It is yet another object of this invention to provide peptides derived from a gp100 melanoma antigen sequence which have been modified to increase their immunogenicity or enhance induction of antimelanoma immune response by enhancing binding to MHC molecules for use in the prophylactic and therapeutic methods described herein
  • It is yet another object of this invention to provide a method of prophylactic or therapeutic immunization for melanoma using the vaccines described herein.
  • It is a further object of this invention to provide a method of identifying melanoma antigens that would constitute potential targets for immunotherapy.
  • It is yet another object of this invention to provide a method of identifying candidate immunogenic peptides derived from either the MART-1 or gp100 sequences for use in immunotherapy.
  • DESCRIPTION OF THE FIGURES
  • FIG. 1 shows the nucleotide and predicted amino acid sequence of the cDNA encoding the MART-1 antigen. The hydrophobic region is underlined.
  • FIG. 2 shows titration of MART-1 peptides for recognition by TIL. T2 cells were incubated with varied concentrations of the purified MART-1 peptides, M9-1, M9-2, M9-3, M10-2, M10-3, M10-4 and M10-5, and lysis by TIL clone A42 or TIL line TIL1235 was measured by 4 h-51Cr release cytotoxicity assay at an E (EFFECTOR):T (TARGET) ratio of 20:1 for A42 and 40:1 for TIL1235. Peptide M9-2 sensitized T2 cells at a concentration of 1 ng/ml. The purified peptide M10-4 was recognized by TIL1235, but not by A42. (M9-1 |-|, M9-2 -, M9-3, ▪-▪, M10-2 ▴-▴, M10-3 ▾-▾, M10-4 -, M10-5 +-+).
  • FIG. 3A shows a radionuclide scan of patient 1200 with metastatic melanoma after receiving the adoptive transfer of autologous 111In labeled TIL1200. The arrow indicates one of the areas of TIL accumulation corresponding to a metastatic lesion in the left thigh.
  • FIG. 3B shows regression of subcutaneous metastatic tumors following treatment with TIL1200 plus IL-2. Treatment began on day 0.
  • FIG. 4 show the nucleic acid sequence of the full length cDNA25. The start and stop codons are underlined.
  • FIG. 5A shows amino acid sequence of the full length cDNA25. The antigenic peptide is underlined.
  • FIG. 5B shows comparison of the amino acid sequence of the full length cDNA25 (cDNA25FL), the truncated form of cDNA25 (cDNA25TR), Pmel17, ME20 and gp100. (•indicates deletion; - indicates identity).
  • FIG. 6 shows northern blot analysis of melanoma and neonatal melanocyte cell lines and various fresh tissues (10-20 μg of total RNA) with a cDNA25 probe(the Sal I digested fragment of pCRII-cDNA25) and the β-actin probe(Clontech). C32, 586mel melanoma cell lines and NHEM529, NHEM530 neonatal melanocyte cell lines were very weak positive.
  • FIGS. 7A-7B show the location of gp100 epitopes and the DNA fragments tested for epitope analysis and recognition by CTL. FIG. 7A. Five DNA fragments (D3, D5, D4, C4, 25TR) tested for epitope analysis are shown ( - - - , identical amino acid). Locations of the identified epitopes are underlined. FIG. 7B. Recognition by CTL (620-1, 620-2, 660-1, 1143, 1200) of COS 7 cells transfected with each DNA fragment in pcDNA3 plasmid along with HLA-A2.1 cDNA by IFN-γ secretion assays are shown (+, recognized; −, not recognized).
  • FIGS. 8A-8D show titration of gp100 epitopes by sensitization of HLA-A2.1+T2 cells for CTL lysis. Lysis of T2 cells preincubated with peptides was tested in a 4 h 51Cr release cytotoxicity assay. FIG. 8A, Lysis by TIL1200 of T2 cells incubated with G9154(▪) or G10 154(). FIG. 8B. Lysis by TIL620 of T2 cells incubated with G9209(▪) or G10208() FIG. 8C. Lysis by TIL660-1 of T2 cells incubated with G9280(▪) FIG. 8D. Lysis by TIL660-2 of T2 cells incubated with G10-5(▪).
  • DETAILED DESCRIPTION OF THE INVENTION
  • For the purpose of a more complete understanding of the invention, the following definitions are described herein. Nucleic acid sequences includes, but is not limited to, DNA, RNA or cDNA. Nucleic acid sequence as used herein refers to an isolated and purified nucleic acid sequence. MART-1 messenger RNA (mRNA) refers to one or more RNA transcripts which are a product of the MART-1 gene. Substantially homologous as used herein refers to substantial correspondence between the nucleic acid sequence of MART-1 shown in FIG. 1 (SEQ ID NO:1) and that of any other nucleic acid sequence. Substantially homologous means about 50-100% homologous homology, preferably by about 70-100% homology, and most preferably about 90-100% homology between the MART-1 sequence and that of any other nucleic acid sequence. In addition, substantially homologous as used herein also refers to substantial correspondences between the amino acid sequence of the MART-1 antigen shown in FIG. 1 (SEQ ID NO:2) and that of any other amino acid sequence.
  • Major Histocompatibility Complex (MHC) is a generic designation meant to encompass the histo-compatibility antigen systems described in different species including the human leucocyte antigens (HLA).
  • The term melanoma includes, but is not limited to, melanomas, metastatic melanomas, melanomas derived from either melanocytes or melanocytes related nevus cells, melanocarcinomas, melanoepitheliomas, melanosarcomas, melanoma in situ, superficial spreading melanoma, nodular melanoma, lentigo maligna melanoma, acral lentiginous melanoma, invasive melanoma or familial atypical mole and melanoma (FAM-M) syndrome. Such melanomas in mammals may be caused by, chromosomal abnormalities, degenerative growth and developmental disorders, mitogenic agents, ultraviolet radiation (UV), viral infections, inappropriate tissue expression of a gene, alterations in expression of a gene, or carcinogenic agents. The aforementioned melanomas can be diagnosed, assessed or treated by methods described in the present application.
  • By atypical mole we mean a mole with features that are abnormal and may be precancerous.
  • By melanoma antigen or immunogen we mean all or parts thereof of the MART-1 protein or peptides based on the MART-1 protein sequence capable of causing a cellular or humoral immune response in a mammal. Such antigens may also be reactive with antibodies from animals immunized with all, part or parts of the MART-1 protein (SEQ ID NO:2). Such a protein or peptide may be encoded by all or part of the MART-1 nucleic acid sequence of this invention.
  • By immunogenic peptide we mean a peptide derived from the MART-1 protein sequence (FIG. 1; SEQ ID NO. 2) or a gp100 protein sequence (FIG. 5A; SEQ ID NO: 27) capable of causing a cellular or humoral immune response in a mammal. Such peptides may be reactive with antibodies from an animal immunized with the peptides. Such peptides are about 5-20 amino acid in length preferably about 8 to 15 amino acids in length, and most preferably about 9-10 amino acids in length.
  • One skilled in the art will understand that the bioassays of the present invention may be used in the analysis of biological samples or tissues from any vertebrate species. In a preferred embodiment, mammalian biological samples or tissues are analyzed.
  • Tissue includes, but is not limited to, single cells, whole organs and portions thereof. Biological samples include, but are not limited to, tissues, primary cultures of mammalian tissues, biopsy specimens, pathology specimens, and necropsy specimens. Mammal includes but is not limited to, humans, monkeys, dogs, cats, mice, rats, pigs, cows, pigs, horses, sheep and goats.
  • The present invention provides a nucleic acid sequence which encodes a novel melanoma antigen recognized by T cells. This novel melanoma antigen designated MART-1 (melanoma antigen-recognized by T-Cells-1). MART-1 shows no significant homology to any known melanoma antigen and thus represents a new melanoma antigen. The MART-1 antigen contains a highly hydrophobic region from amino acids 27 to 47 (SEQ ID. NO:2) followed by three arginine residues, suggestive of a transmembrane protein. Although no significant homology exists to the entire protein there is a 27 amino acid segment (amino acids 57-83; SEQ ID. NO:2) that is 37% identical to a Type II membrane protein previously recognized as mouse natural killer cell surface protein NKR-P1 (Yokoyama, W. M., et al. (1991), J. Immunol. 147:3229-3236). MART-1 does not contain a leader sequence characteristic of many Type I membrane proteins (Singer, S. J. (1990) Annu. Rev. Cell Biol. 6: 247-296).
  • MART-1 RNA expression appears to be restricted to fresh and cultured melanoma and melanocyte cell lines and human retina; expression has not been found in any other fresh or cultured tissues or other tumor histologies tested. The cDNA sequence for MART-1 is shown in FIG. 1 (SEQ ID NO:1), the deduced amino acid sequence for the MART-1 protein is also shown in FIG. 1 (SEQ ID NO.:1).
  • The nucleic acid sequence for MART-1 shown in FIG. 1 (SEQ ID NO.:1), represents a preferred embodiment of the invention. It is, however, understood by one skilled in the art that due to the degeneracy of the genetic code variations in the cDNA sequence shown in FIG. 1 (SEQ ID NO.:1) will still result in a DNA sequence capable of encoding the MART-1 protein antigen. Such DNA sequences are therefore functionally equivalent to the sequence set forth in FIG. 1 (SEQ ID NO.:1) and are intended to be encompassed within the present invention. Further, a person of skill in the art will understand that there are naturally occurring allelic variations in a given species of the MART-1 nucleic acid sequence shown in FIG. 1 (SEQ ID NO.:1), these variations are also intended to be encompassed by the present invention.
  • The predicted MART-1 antigen is a 118 amino acid protein of about 13 (kd). This invention further includes MART-1 protein or peptides or analogs thereof having substantially the same function as the MART-1 antigen or protein of this invention. Such proteins or polypeptides include, but are not limited to, a fragment of the protein, or a substitution, addition or deletion mutant of the MART-1 protein. This invention also encompasses proteins or peptides that are substantially homologous to the MART-1 antigen. Substantially homologous means about 50-100% homology, preferably about 70-100% homology, and most preferably about 90-100% homology between the MART-1 and any another amino acid sequence or protein or peptide.
  • The term “analog” includes any polypeptide having an amino acid residue sequence substantially identical to the MART-1 sequence specifically shown herein (FIG. 1; SEQ ID NO: 1) in which one or more residues have been conservatively substituted with a functionally similar residue and which displays the functional aspects of the MART-1 antigen as described herein. Examples of conservative substitutions include the substitution of one non-polar (hydrophobic) residue such as isoleucine, valine, leucine or methionine for another, the substitution of one polar (hydrophilic) residue for another such as between arginine and lysine, between glutamine and asparagine, between glycine and serine, the substitution of one basic residue such as lysine, arginine or histidine for another, or the substitution of one acidic residue, such as aspartic acid or glutamic acid or another.
  • The phrase “conservative substitution” also includes the use of a chemically derivatized residue in place of a non-derivatized residue. “Chemical derivative” refers to a subject polypeptide having one or more residues chemically derivatized by reaction of a functional side group. Examples of such derivatized molecules include for example, those molecules in which free amino groups have been derivatized to form amine hydrochlorides, p-toluene sulfonyl groups, carbobenzoxy groups, t-butyloxycarbonyl groups, chloroacetyl groups or formyl groups. Free carboxyl groups may be derivatized to form salts, methyl and ethyl esters or other types of esters or hydrazides. Free hydroxyl groups may be derivatized to form O-acyl or O-alkyl derivatives. The imidazole nitrogen of histidine may be derivatized to form N-im-benzylhistidine. Also included as chemical derivatives are those proteins or peptides which contain one or more naturally-occurring amino acid derivatives of the twenty standard amino acids. For examples: 4-hydroxyproline may be substituted for proline; 5-hydroxylysine may be substituted for lysine; 3-methylhistidine may be substituted for histidine; homoserine may be substituted for serine; and ornithine may be substituted for lysine. Proteins or polypeptides of the present invention also include any polypeptide having one or more additions and/or deletions or residues relative to the sequence of a polypeptide whose sequence is encoded is the DNA of MART-1, so long as the requisite activity is maintained.
  • This invention also provides a recombinant DNA molecule comprising all or part of the MART-1 nucleic acid sequence (SEQ. ID NO.:1) and a vector. Expression vectors suitable for use in the present invention may comprise at least one expression control element operationally linked to the nucleic acid sequence. The expression control elements are inserted in the vector to control and regulate the expression of the nucleic acid sequence. Examples of expression control elements include, but are not limited to, lac system, operator and promoter regions of phage lambda, yeast promoters and promoters derived from polyoma, adenovirus, retrovirus or SV40. Additional preferred or required operational elements include, but are not limited to, leader sequence, termination codons, polyadenylation signals and any other sequences necessary or preferred for the appropriate transcription and subsequent translation of the nucleic acid sequence in the host system. It will be understood by one skilled in the art the correct combination of required or preferred expression control elements will depend on the host system chosen. It will further be understood that the expression vector should contain additional elements necessary for the transfer and subsequent replication of the expression vector containing the nucleic acid sequence in the host system. Examples of such elements include, but are not limited to, origins of replication and selectable markers. It will further be understood by one skilled in the art that such vectors are easily constructed using conventional methods (Ausubel et al., (1987) in “Current Protocols in Molecular Biology”, John Wiley and Sons, New York, N.Y.) or commercially available.
  • Another aspect of this invention relates to a host organism into which recombinant expression vector containing all or part of the MART-1 nucleic acid sequence has been inserted. The host cells transformed with the MART-1 nucleic acid sequence of this invention include eukaryotes, such as animal, plant, insect and yeast cells and prokaryotes, such as E. coli. The means by which the vector carrying the gene may be introduced into the cell include, but are not limited to, microinjection, electroporation, transduction, or transfection using DEAE-dextran, lipofection, calcium phosphate or other procedures known to one skilled in the art (Sambrook et al. (1989) in “Molecular Cloning. A Laboratory Manual”, Cold Spring Harbor Press, Plainview, N.Y.).
  • In a preferred embodiment, eukaryotic expression vectors that function in eukaryotic cells are used. Examples of such vectors include, but are not limited to, retroviral vectors, vaccinia virus vectors, adenovirus vectors, herpes virus vector, fowl pox virus vector, bacterial expression vectors, plasmids, such as pcDNA3 (Invitrogen, San Diego, Calif.) or the baculovirus transfer vectors. Preferred eukaryotic cell lines include, but are not limited to, COS cells, CHO cells, HeLa cells, NIH/3T3 cells, 293 cells (ATCC#CRL1573), T2 cells, dendritic cells, or monocytes. In a particularly preferred embodiment the recombinant MART-1 protein expression vector is introduced into mammalian cells, such as NIH/3T3, COS, CHO, 293 cells (ATCC #CRL 1573), T2 cells, dendritic cells, or monocytes to ensure proper processing and modification of the MART-1 protein. In an alternative embodiment the MART-1 cDNA is introduced into COS7 (Gluzman, Y. et al. (1981) Cell 23: 175-182). The choice of an appropriate cell is within the skill of a person in the art.
  • In one embodiment the expressed recombinant MART-1 protein may be detected by methods known in the art which include Coomassie blue staining and Western blotting using antibodies specific for the MART-1 protein.
  • In a further embodiment, the recombinant protein expressed by the host cells can be obtained as a crude lysate or can be purified by standard protein purification procedures known in the art which may include differential precipitation, molecular sieve chromatography, ion-exchange chromatography, isoelectric focusing, gel electrophoresis, affinity, and immunoaffinity chromatography and the like. (Ausubel et. al., (1987) in “Current Protocols in Molecular Biology” John Wiley and Sons, New York, N.Y.). In the case of immunoaffinity chromatography, the recombinant protein may be purified by passage through a column containing a resin which has bound thereto antibodies specific for the MART-1 protein (Ausubel et. al., (1987) in “Current Protocols in Molecular Biology” John Wiley and Sons, New York, N.Y.).
  • The nucleic acid sequence or portions thereof, of this invention are useful as probes for the detection of expression of the MART-1 gene in normal and diseased tissue. Therefore, another aspect of the present invention relates to a bioassay for detecting messenger RNA encoding the MART-1 protein in a biological sample comprising the steps of (a) contacting all or part of the nucleic acid sequence of this invention with said biological sample under conditions allowing a complex to form between said nucleic acid sequence and said messenger RNA, (b) detecting said complexes and, (c) determining the level of said messenger RNA.
  • RNA can be isolated as whole cell RNA or as poly(A)+ RNA. Whole cell RNA can be isolated by a variety of methods known to those skilled in the art. (Ausubel et al., (1987) on “Current Protocols in Molecular Biology”, John Wiley and Sons, New York). Such methods include extraction of RNA by differential precipitation (Birnboim, H. C. (1988) Nucleic Acids Res., 16:1487-1497), extraction of RNA by organic solvents (Chomczynski, P. et al. (1987) Anal. Biochem., 162:156-159) and the extraction of RNA with strong denaturants (Chirgwin, J. M. et al. (1979) Biochemistry, 18:5294-5299). Poly(A)+ RNA can be selected from whole cell RNA by affinity chromatography on oligo-d(T) columns (Aviv, H. et al. (1972) Proc. Natl. Acad. Sci., 69:1408-1412). Examples of methods for determining cellular messenger mRNA levels for step (c) include, but are not limited to Northern blotting (Alwine, J. C. et al. (1977) Proc. Natl. Acad. Sci., 74:5350-5354), dot and slot hybridization (Kafatos, F. C. et al. (1979) Nucleic Acids Res., 7:1541-1522), filter hybridization (Hollander, M. C. et al. (1990) Biotechniques; 9:174-179), RNase protection (Sambrook et. al., (1989) in “Molecular Cloning, A Laboratory Manual”, Cold Spring Harbor Press, Plainview, N.Y.), polymerase chain reaction (Watson, J. D. et al. (1992) in “Recombinant DNA” Second Edition, W. H. Freeman and Company, New York) and nuclear run-off assays (Ausubel et. al., (1987) in “Current Protocols in Molecular Biology” Supplement 9 (1990); John Wiley and Sons, New York, N.Y.).
  • Detection of complexes in Step (b) of the bioassay can also be carried out by a variety of techniques. Detection of the complexes by signal amplification can be achieved by several conventional labelling techniques including radiolabels and enzymes (Sambrook et. al., (1983) in “Molecular Cloning, A Laboratory Manual”, Cold Spring Harbor Press, Plainview, N.Y.; Ausubel et al., (1987) in “Current Protocols in Molecular Biology, John Wiley and Sons, New York N.Y.). Radiolabelling kits are also commercially available. The MART-1 nucleic acid sequence used as a probe in step(c) of the bioassay may be RNA or DNA. Preferred methods of labelling the DNA sequences are with 32P using Klenow enzyme or polynucleotide kinase. Preferred methods of labeling RNA or riboprobe sequences are with 32P or 35S using RNA polymerases. In addition, there are known non-radioactive techniques for signal amplification including methods for attaching chemical moieties to pyrimidine and purine rings (Dale, R. N. K. et al. (1973) Proc. Natl. Acad. Sci., 70:2238-2242; Heck, R. F. (1968) S. Am. Chem. Soc., 90:5518-5523), methods which allow detection by chemiluminescence (Barton, S. K. et al. (1992) J. Am. Chem. Soc., 114:8736-8740) and methods utilizing biotinylated nucleic acid probes (Johnson, T. K. et al. (1983) Anal. Biochem., 133:125-131; Erickson, P. F. et al. (1982) J. of Immunology Methods, 51:241-249; Matthaei, F. S. et al (1986) Anal. Biochem., 157:123-128) and methods which allow detection by fluorescence using commercially available products. Non-radioactive labelling kits are also commercially available.
  • Examples of biological samples that can be used in this bioassay include, but are not limited to, primary mammalian cultures, continuous mammalian cell lines, such as melanocyte cell lines, mammalian organs such as skin or retina, tissues, biopsy specimens, neoplasms, pathology specimens, and necropsy specimens.
  • In a preferred embodiment, a 32P radiolabelled MART-1 probe, as exemplified in Example 1, is used. Preferably the MART-1 probe is the full length cDNA in FIG. 1 (SEQ ID NO:1). The approximately 1.6 Kilobase (kb) cDNA (FIG. 1; SEQ ID NO:1) was cloned into the vector and the resulting plasmid deposited with the American Type Culture Collection (ATCC) 12301 Parklawn Drive, Rockville, Md. 20852 USA on Apr. 14, 1994, and given ATCC Deposit Number 75738. The full length MART-1 nucleic acid sequence can be isolated from the pCRII plasmid by digestion with HINDIII and XhoI restriction enzymes. This 1.6 kb nucleic acid sequence can then be used as a probe. This probe is used to detect MART-1 mRNA in total RNA or poly A+ RNA isolated from a variety of tissues or biological samples.
  • In another embodiment, combinations of oligonucleotide pairs based on the MART-1 sequence in FIG. 1 (SEQ ID NO.:1) are used as Polymerase Chain Reaction (PCR) primers to detect MART-1 mRNA in a biological sample. These primers can be used in a method following the reverse transcriptase-Polymerase Chain Reaction (RT-PCR) process for amplifying selected RNA nucleic acid sequences as detailed in Ausubel et al., (eds) (1987) In “Current Protocols in Molecular Biology” Chapter 15, John Wiley and Sons, New York, N.Y. The oligonucleotides can be synthesized by automated instruments sold by a variety of manufacturers or can be commercially prepared based upon the nucleic acid sequence of this invention. One skilled in the art will know how to select PCR primers based on the MART-1 nucleic acid sequence for amplifying MART-1 RNA in a sample.
  • The MART-1 nucleic acid sequence or portions thereof (FIG. 1: SEQ ID NO:1) of this invention are useful to detect alterations of the MART-1 gene in normal or diseased mammalian tissue. By alteration, we mean additions, deletions, substitutions or duplications in the MART-1 gene sequence or gene amplification of the MART-1 gene sequence. Therefore, another aspect of the present invention relates to an assay for detecting alterations of the MART-1 gene in a biological sample comprising the steps of (a) contacting all or part of the nucleic acid sequence of this invention with genomic DNA isolated from a biological sample under conditions allowing a complex to form between said nucleic acid sequence and said genomic DNA, (b) detecting said complexes, and (c) determining alterations in said MART-1 gene by comparison to a control sample.
  • Standard methods for isolating DNA from a biological sample, detecting alterations in a gene and detecting complex between the MART-1 nucleic acid probe and genomic DNA sequences are provided in manuals such as Sambrook et al., (eds) (1989) “Molecular Cloning, A Laboratory Mineral”, Cold Spring Harbor Press, Plainview, N.Y. and in Ausubel et al., (eds) (1987) in “Current Protocols in Molecular Biology” John Wiley and Sons, New York, N.Y.
  • The MART-1 nucleic acid sequences of this invention (FIG. 1; SEQ ID No:1) can also be used as probes to isolate the MART-1 homologs in other species. In a preferred embodiment the MART-1 cDNA (FIG. 1; SEQ ID No:1) is used to screen a mammalian cDNA library, positive clones are selected and sequenced. Examples of tissue sources from which the cDNA library can be synthesized include, but are not limited to skin, retina, melanocytes, neonatal skin and embryos. Preferably a melanoma library is screened using the MART-1 cDNA as a probe (FIG. 1; SEQ ID No. 1). One skilled in the art will understand the appropriate hybridization conditions to be used to detect the homologs. Conventional methods for nucleic acid hybridization, construction of libraries and cloning techniques are described in Sambrook et al., (eds) (1989) In “Molecular Cloning A Laboratory Manual” Cold Spring Harbor Press, Plainview, N.Y. and Ausubel et al., (eds) in “Current Protocols in Molecular Biology” (1987), John Wiley and Sons, New York, N.Y.
  • It is known that all or parts thereof of the MART-1 protein is an antigen present on melanoma cells. It is therefore another aspect of this invention to provide MART-1 nucleic acid probes to be utilized in detecting MART-1 RNA or alterations in the level of MART-1 mRNA in biological sample isolated from a mammal afflicted with a disease. Examples of such diseases, include but are not limited to, melanomas. By alterations in the level of MART-1 mRNA we mean an increase or decrease in the level of an RNA relative to a control sample or the appearance or disappearance of the MART-1 mRNA relative to a control sample. Detection in the alterations of MART-1 mRNA will allow for diagnosis or the assessment of the diseased state. Therefore, alterations in the level of MART-1 mRNA may be predictive of the prognosis for the afflicted mammal.
  • In another embodiment the nucleic acid of this invention can be used in in situ hybridization on mammalian tissues to determine the precise site or subcellular site of expression of the MART-1 gene within a tissue. A preferred method of labeling the MART-1 nucleic acid sequence is synthesizing a 35S-labeled RNA probe by in vitro transcription utilizing SP6 polymerase. In the MART-1 plasmid (ATCC Deposit #75738) the sense strand is under the control of the T7 promoter, the antisense strand is under the SP6 promoter. It is preferable that the probe be hydrolyzed to a probe length of approximately 400-200 base pairs. Conventional methods for preparation of tissues for in situ, synthesis of probes and detection of signal can be found in Ausubel et. al., (eds) (1987) in “Current Protocols in Molecular Biology,” John Wiley and Sons, New York, N.Y. Chapter 14 and Vander Ploeg, M., Raap A. K. (1988) In “New Frontiers in Cytology” Goerttler, K., Feichter, G E, Witte. S. (eds) pp 13-21 Springer-Verlag, New York. The probe is then contacted with mammalian tissue sections and in situ analyses performed by conventional methods. Examples of tissues that can be used include, but are not limited to, mammalian embryos, adult mammalian tissues, such as skin, lymph nodes and retina, biopsy specimens, pathology specimens and necropsy specimens. In a preferred embodiment, MART-1 in situ probes may be used to evaluate MART-1 RNA expression in diseased tissue for invasive early melanoma to characterize radial and vertical growth phases of the melanoma lesion and assess the margins of the disease within the tissue.
  • In yet another embodiment of this invention all or parts thereof of the MART-1 (SEQ ID NO:1) nucleic acid sequence can be used to generate transgenic animals. Preferably the MART-1 gene is introduced into an animal or an ancestor of the animal at an embryonic stage, preferably at the one cell stage and generally not later than about the eight cell stage. There are several means by which transgenic animals carrying a MART-1 gene can be made. One method involves the use of retroviruses carrying all or part of the MART-1 sequence. The retroviruses containing the transgene are introduced into the embryonic animal by transfection. Another methods involves directly injecting the transgene into the embryo. Yet another methods employs the embryonic stem cell method or homologous recombination method known to workers in the field. Examples of animals into which the MART-1 transgene can be introduced include but are not limited to, primates, mice, rats or other rodents. Such transgenic animals may be useful as biological models for the study of melanoma and to evaluate diagnostic or therapeutic methods for melanoma.
  • This invention further comprises an antibody or antibodies reactive with the MART-1 protein or peptides or modified peptides or analogs thereof having the amino acid sequence defined in FIG. 1 (SEQ ID NO: 2) or a unique portion thereof. In this embodiment of the invention the antibodies are monoclonal or polyclonal in origin. MART-1 protein or peptides used to generate the antibodies may be from natural or recombinant sources or generated by chemical synthesis. Natural MART-1 proteins can be isolated from mammalian biological samples. Biological samples include, but are not limited to mammalian tissues such as fresh melanoma, skin, retina, primary or continuous cultures of mammalian cells such as melanoma cultures or cultured melanocytes. The natural MART-1 proteins may be isolated by the same methods described above for recombinant proteins. Recombinant MART-1 proteins or peptides may be produced and purified by conventional methods. Synthetic MART-1 peptides may be custom ordered or commercially made based on the predicted amino acid sequence of the present invention (FIG. 1; SEQ ID:2) or synthesized by methods known to one skilled in the art (Merrifield, R. B. (1963) J. Amer. Soc. 85:2149). Examples of MART-1 peptides include, but are not limited to, are AAGIGILTV (M9-2; SEQ. ID NO. 4), EAAGIGILTV (M10-3; SEQ. ID NO. 17) and AAGIGILTVI (M10-4; SEQ. ID NO. 18) (peptides are presented in single letter amino acid code). The most preferred peptide is AAGIGILTV (SEQ ID NO:4).
  • Alternatively, peptides derived form the MART-1 protein sequence may modified to increase their immunogenicity by enhancing binding of the peptide to the MHC molecules in which the peptide is presented. Examples of such modified MART-1 peptides that may used are shown, but not limited to, the peptides in Table 14. In a preferred embodiment the MART-1 peptide that is modified to enhance its binding to MHC Class I molecules is AAGIGILTV (SEQ ID NO:4). By way of example, the modified peptides ALGIGILTV (M9-2-2L), WAGIGILTV (M9-2-1W), FAGIGILTV (M9-2-1F) and AAYIGILTV (M9-2-3Y). The peptide or modified peptide may be conjugated to a carrier molecule to enhance the antigenicity of the peptide. Examples of carrier molecules, include, but are not limited to, human albumin, bovine albumin, lipoprotein and keyhole limpet hemo-cyanin (“Basic and Clinical Immunology” (1991) Stites, D. P. and Terr A. I. (eds) Appleton and Lange, Norwalk Connecticut, San Mateo, Calif.).
  • Exemplary antibody molecules for use in the detection methods of the present invention are intact immunoglobulin molecules, substantially intact immunoglobulin molecules or those portions of an immunoglobulin molecule that contain the antigen binding site, including those portions of immunoglobulin molecules known in the art as F(ab), F(ab′); F(ab)2 and F(v). Polyclonal or monoclonal antibodies may be produced by methods known in the art. (Kohler and Milstein (1975) Nature 256, 495-497; Campbell “Monoclonal Antibody Technology, the Production and Characterization of Rodent and Human Hybridomas” in Burdon et al. (eds.) (1985) “Laboratory Techniques in Biochemistry and Molecular Biology,” Volume 13, Elsevier Science Publishers, Amsterdam). The antibodies or antigen binding fragments may also be produced by genetic engineering. The technology for expression of both heavy and light chain genes in E. coli is the subject of the PCT patent applications: publication number WO 901443, WO 901443 and WO 9014424 and in Huse et al. (1989) Science 246:1275-1281.
  • The antibodies of this invention may react with native or denatured MART-1 protein, peptides or analogs thereof, or modified peptides an analogs thereof. The specific immunoassay in which the antibodies are to be used will dictate which antibodies are desirable. Antibodies may be raised against the MART-1 protein or portions thereof or against synthetic peptides homologous to the MART-1 amino acid sequence.
  • In one embodiment the antibodies of this invention are used in immunoassays to detect the novel MART-1 protein in biological samples. In this method the antibodies of the present invention are contacted with a biological sample and the formation of a complex between the MART-1 antigen and antibody is detected. Immunoassays of the present invention may be radioimmunoassay, Western blot assay, immunofluorescent assay, enzyme immunoassay, chemiluminescent assay, immunohistochemical assay and the like. (In “Principles and Practice of Immunoassay” (1991) Christopher P. Price and David J. Neoman (eds), Stockton Press, New York, N.Y.; Ausubel et al. (eds) (1987) in “Current Protocols in Molecular Biology” John Wiley and Sons, New York, N.Y.). Standard techniques known in the art for ELISA are described in Methods in Immunodiagnosis, 2nd Edition, Rose and Bigazzi, eds., John Wiley and Sons, New York 1980 and Campbell et al., Methods of Immunology, W. A. Benjamin, Inc., 1964, both of which are incorporated herein by reference. Such assays may be direct, indirect, competitive, or noncompetitive immunoassays as described in the art (In “Principles and Practice of Immunoassay” (1991) Christopher P. Price and David J. Neoman (eds), Stockton Pres, NY, N.Y.; Oellirich, M. 1984. J. Clin. Chem. Clin. Biochem. 22: 895-904) Biological samples appropriate for such detection assays include mammalian tissues, melanoma and melanocyte cell lines, skin, retina, lymph nodes, pathology specimens, necropsy specimens, and biopsy specimens. Proteins may be isolated from biological samples by conventional methods described in (Ausubel et al., (eds) (1987) in “Current Protocols in Molecular Biology” John Wiley and Sons, New York, N.Y.).
  • The antibodies of this invention can therefore be used in immunoassays to detect MART-1 antigen or alteration in the level of expression of the MART-1 antigen in biological samples isolated from mammals afflicted with a disease or disorder. Examples of biological samples include, but are not limited to, mammalian tissues, biopsy tissue samples, melanoma and lymph node biopsy samples, pathology and tissue samples. Examples of diseases that can be assessed by these immunoassays, include, but are not limited to, melanomas and tissues which are secondary sites for melanoma metastasis. By alteration in level of expression, we mean an increase or decrease of the MART protein or portions thereof relative to a control sample. Alteration is also meant to encompass substitution, deletion or addition mutants of the MART-1 protein. Such mutations can be determined by using the antibodies of this invention known to react with specific epitopes of the MART-1 protein and determining which epitopes are present relative to a control. The antibodies of this invention can therefore be used in an immunoassay to diagnose, assess or prognoses a mammal afflicted with the disease.
  • In a preferred embodiment, the MART-1 antibodies of this invention are used to assess the presence of the MART-1 antigen from a tissue biopsy of a mammal afflicted with melanoma using immunocytochemistry. Such assessment of the delineation of the MART-1 antigen in a diseased tissue can be used to prognose the progression of the disease in a mammal afflicted with the disease. Specifically the MART-1 antibodies can be used to characterize the radial and vertical growth phases of the melanoma lesion. Conventional methods for immunohistochemistry are described in (Harlow and Lane (eds) (1988) In “Antibodies A Laboratory Manual”, Cold Spinning Harbor Press, Cold Spring Harbor, N.Y.; Ausbel et al. (eds) (1987). In Current Protocols In Molecular Biology John Wiley and Sons (New York, N.Y.).
  • In another embodiment, antibodies of this invention may be used to purify the MART-1 protein or portions thereof. Immunoaffinity chromatography can be performed by conventional methods known to one skilled in the art (Ausubel et al. (eds) (1987) in “Current Protocols in Molecular Biology” John Wiley and Sons, New York, N.Y.).
  • In another preferred embodiment rabbit antisera containing antibodies which specifically recognize the MART-1 protein is used to detect said protein in Western Blot Analysis. Such antisera is directed to all, or a part or parts of the MART-1 protein or synthetic peptides derived from the MART-1 protein sequence. Preferably a MART-1 synthetic peptide derived from the MART-1 predicted amino acid sequence is used (FIG. 1; SEQ ID NO:2). Alternatively, modified MART-1 peptides may be used. The peptide is synthesized by standard methods on an automated peptide synthesizer and purified by high pressure liquid chromatography (HPLC) as described in Example 2. The purified peptide may be conjugated to a carrier as described in (M. Bodanszky (1984) “Principles of Peptide Synthesis,” Springer Verlag, New York, N.Y.). Using conventional methods, rabbits may be immunized with the MART-1 protein or peptide conjugated to carriers. Preferably about 0.1 to about 10 (mg) of antigen in adjuvant may be used, most preferably about 1 mg of antigen in adjuvant may be used. The animal receives similar booster doses and antisera titer is assessed by ELISA assay. Satisfactory levels of antisera are obtained when the anti-peptide antibody titer reaches a plateau. This antibody can be used in the standard immunoassays described above.
  • T-lymphocytes recognize antigen in association with Class I or Class II MHC molecules in the form of a peptide fragment bound to an MHC molecule. The degree of peptide binding to a given MHC allele is based on amino acids at particular positions within the peptide (Parker et al. (1992) Journal of Immunology 149:3580; Kubo, et al. (1994) Journal of Immunology 52:3913-3924; Ruppert J. et al. (1993) Cell 74:929-937; Falk et al. (1991) Nature 351:290-296, each of which is herein incorporated by reference). Therefore, another embodiment of this invention relates to peptides derived from the MART-1 protein sequence (FIG. 1; SEQ ID NO:2) which have been modified to increase immunogenicity by enhancing binding of the peptide to the MHC molecule with which the peptide is associated. By way of example, modification may include substitution, deletion or addition of an amino acid in the given-immunogenic peptide sequence or mutation of existing amino acids within the given immunogenic peptide sequence, or derivatization of existing amino acids within the given immunogenic peptide sequence. Any amino acid comprising the immunogenic peptide sequence may be modified in accordance with this invention. In a preferred embodiment at least one amino acid is substituted or replaced within the given immunogenic peptide sequence. Any amino acid may be used to substitute or replace a given amino acid within the immunogenic peptide sequence. Modified peptides are intended to include any immunogenic MART-1 peptide which has been modified and exhibits enhanced binding to the MHC molecule with which it associates when presented to the T-cell.
  • By way of example, the HLA-A2 allele binds peptides of nine or ten amino acids. Examples of positions within the peptide that may be altered to enhance binding include, but are not limited to, the first position, the second position, the third position and the last position of the peptide. Any amino acid may be used to substitute or replace these positions within the immunogenic peptide sequence. For enhanced binding to HLA-A2 the amino acid at the second position of the peptide is preferably a hydrophobia aliphatic amino acid. Examples or amino acids that may be used at the second position include, but are not limited to, leucine, methionine, alanine, isoleucine, valine, threonine or glycine. Preferably leucine or methionine is found at the second position of the peptide. The last amino acid of the peptide (either the 9th or 10th amino acid depending on the length of the peptide) is preferably a hydrophobic aliphatic amino acid. Examples of amino acids that may be used in the last position of the peptide include, but are not limited to, valine, methionine, leucine, alanine, isoleucine, threonine or glycine. Preferably valine is found at the last position in the peptide. The amino acids at the first and third positions in the peptide may also be modified to enhance binding of the peptide to the MHC Class I molecule. The amino acids at the first and third positions in the peptide may be any amino acid. Preferably, the amino acids at the first and third positions are hydrophobic aliphatic amino acids or aromatic amino acids. Examples of amino acids that maybe used at these positions include, but are not limited to, leucine, methionine, valine, alanine, isoleucine, threonine, glycine, tryptophan, phenylalanine, tyrosine, serine, aspartic acid, or lysine. Examples of MART-1 peptides that may be modified include, but are not limited to, AAGIGILTV (SEQ ID NO: 4), EAAGIGILTV (SEQ ID NO: 17) and AAGIGILTVI (SEQ ID NO: 18) (peptides are presented in single letter amino acid code). By way of example the immunogenic MART-1 peptide AAGIGILTV (SEQ ID NO:4) may be modified according to the following formula X1X2X3IGILTX4 wherein
  • X1 may be any amino acid, preferably any hydrophobic aliphatic amino acid, or aromatic amino acid. Examples of amino acids that may be used, but are not limited to, alanine, tryptophan, phenylalanine, tyrosine, lysine, isoleucine, leucine, methionine, threonine, glycine or serine.
  • X2 may be any hydrophobic amino acid, preferably an aliphatic hydrophobic amino acids. Examples of amino acids that may be used include, but are not limited to, leucine, methionine, isoleucine, valine, threonine, alanine or glycine.
  • X3 may be any amino acid, preferably any hydrophobic aliphatic amino acid, or aromatic amino acid. Examples of amino acids that may be used include, but are not limited to, tryptophan, phenylalanine, tyrosine, lysine, aspartic acid, serine, alanine, glycine, isoleucine, valine, or threonine.
  • X4 may be any hydropbic amino acid, preferably a hydrophobic aliphatic amino acid. Examples of amino acids that may be used include, but are not limited to, valine, leucine, isoleucine, alanine, threonine, or glycine.
  • Examples of modified AAGIGILTV (SEQ ID NO:4) peptide sequences that may be produced are shown but not limited to the peptides in Table 14 (Example 5).
  • This invention further includes analogs of these immunogenic modified peptides derived from the MART-1 amino acid sequence (FIG. 1; SEQ ID NO:2) which have been modified. The term analog is intended to include any peptide which displays the functional aspects of these modified peptides. The term analog also includes conservative substitutions or chemical derivatives of these modified peptides as described above. These modified peptides may be synthetically or recombinantly produced by conventional methods.
  • The recombinant or natural MART-1 protein, peptides, or analogs thereof, or modified MART-1 peptides, or analogs thereof may be used as a vaccine either prophylactically or therapeutically. When provided prophylactically the vaccine is provided in advance of any evidence of melanoma. The prophylactic administration of the MART-1 vaccine should serve to prevent or attenuate melanoma in a mammal. In a preferred embodiment mammals, preferably human, at high risk for melanoma are prophylactically treated with the vaccines of this invention. Examples of such mammals include, but are not limited to, humans with a family history of melanoma, humans with a history of atypical moles, humans with a history of FAM-M syndrome or humans afflicted with melanoma previously resected and therefore at risk for reoccurrence. When provided therapeutically, the vaccine is provided to enhance the patient's own immune response to the tumor antigen present on the melanoma or metastatic melanoma. The vaccine, which acts as an immunogen, may be a cell, cell lysate from cells transfected with a recombinant expression vector, cell lysates from cells transfected with a MART-1 recombinant expression vector, or a culture supernatant containing the expressed protein. Alternatively, the immunogen is a partially or substantially purified recombinant MART-1 protein, peptide or analog thereof or modified peptides or analogs thereof. The proteins or peptides may be conjugated with lipoprotein or administered in liposomal form or with adjuvant.
  • While it is possible for the immunogen to be administered in a pure or substantially pure form, it is preferable to present it as a pharmaceutical composition, formulation or preparation.
  • The formulations of the present invention, both for veterinary and for human use, comprise an immunogen as described above, together with one or more pharmaceutically acceptable carriers and, optionally, other therapeutic ingredients. The carrier(s) must be “acceptable” in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. The formulations may conveniently be presented in unit dosage form and may be prepared by any method well-known in the pharmaceutical art.
  • All methods include the step of bringing into association the active ingredient with the carrier which constitutes one or more accessory ingredients. In general, the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product into the desired formulation.
  • Formulations suitable for intravenous intramuscular, subcutaneous, or intraperitoneal administration conveniently comprise sterile aqueous solutions of the active ingredient with solutions which are preferably isotonic with the blood of the recipient. Such formulations may be conveniently prepared by dissolving solid active ingredient in water containing physiologically compatible substances such as sodium chloride (e.g. 0.1-2.0M), glycine, and the like, and having a buffered pH compatible with physiological conditions to produce an aqueous solution, and rendering said solution sterile. These may be present in unit or multi-dose containers, for example, sealed ampoules or vials.
  • The formulations of the present invention may incorporate a stabilizer. Illustrative stabilizers are polyethylene glycol, proteins, saccharides, amino acids, inorganic acids, and organic acids which may be used either on their own or as admixtures. These stabilizers are preferably incorporated in an amount of 0.11-10,000 parts by weight per part by weight of immunogen. If two or more stabilizers are to be used, their total amount is preferably within the range specified above. These stabilizers are used in aqueous solutions at the appropriate concentration and pH. The specific osmotic pressure of such aqueous solutions is generally in the range of 0.1-3.0 osmoles, preferably in the range of 0.8-1.2. The pH of the aqueous solution is adjusted to be within the range of 5.0-9.0, preferably within the range of 6-8. In formulating the immunogen of the present invention, anti-adsorption agent may be used.
  • Additional pharmaceutical methods may be employed to control the duration of action. Controlled release preparations may be achieved through the use of polymer to complex or absorb the proteins or their derivatives. The controlled delivery may be exercised by selecting appropriate macromolecules (for example polyester, polyamino acids, polyvinyl, pyrrolidone, ethylenevinylacetate, methylcellulose, carboxymethylcellulose, or protamine sulfate) and the concentration of macromolecules as well as the methods of incorporation in order to control release. Another possible method to control the duration of action by controlled-release preparations is to incorporate the MART-1 protein, peptides and analogs thereof into particles of a polymeric material such as polyesters, polyamino acids, hydrogels, poly(lactic acid) or ethylene vinylacetate copolymers. Alternatively, instead of incorporating these agents into polymeric particles, it is possible to entrap these materials in microcapsules prepared, for example, by coacervation techniques or by interfacial polymerization, for example, hydroxymethylcellulose or gelatin-microcapsules and poly(methylmethacylate) microcapsules, respectively, or in colloidal drug delivery systems, for example, liposomes, albumin microspheres, microemulsions, nanoparticles, and nanocapsules or in macroemulsions.
  • When oral preparations are desired, the compositions may be combined With typical carriers, such as lactose, sucrose, starch, talc magnesium stearate, crystalline cellulose, methyl cellulose, carboxymethyl cellulose, glycerin, sodium alginate or gum arabic among others.
  • The proteins of the present invention may be supplied in the form of a kit, alone, or in the form of a pharmaceutical composition as described above.
  • Vaccination can be conducted by conventional methods. For example, the immunogen can be used in a suitable diluent such as saline or water, or complete or incomplete adjuvants. Further, the immunogen may or may not be bound to a carrier to make the protein immunogenic. Examples of such carrier molecules include but are not limited to bovine serum albumin (BSA), keyhole limpet hemocyanin (KLH), tetanus toxoid, and the like. The immunogen also may be coupled with lipoproteins or administered in liposomal form or with adjuvants. The immunogen can be administered by any route-appropriate for antibody production such as intravenous, intraperitoneal, intramuscular, subcutaneous, and the like. The immunogen may be administered once or at periodic intervals until a significant titer of anti-MART-1 immune cells or anti-MART-1 antibody is produced. The presence of anti-MART-1 immune cells may be assessed by measuring the frequency of precursor CTL (cytoxic T-lymphocytes) against MART-1 antigen prior to and after immunization by a CTL precursor analysis assay (Coulie, P. et al., (1992) International Journal Of Cancer 50:289-297). The antibody may be detected in the serum using the immunoassay described above.
  • The administration of the vaccine or immunogen of the present invention may be for either a prophylactic or therapeutic purpose. When provided prophylactically, the immunogen is provided in advance of any evidence or in advance of any symptom due to melanoma. The prophylactic administration of the immunogen serves to prevent or attenuate melanoma in a mammal. When provided therapeutically, the immunogen is provided at (or shortly after) the onset of the disease or at the onset of any symptom of the disease. The therapeutic administration of the immunogen serves to attenuate the disease.
  • A preferred embodiment is a vaccine prepared using recombinant MART-1 protein or peptide expression vectors. To provide a vaccine to an individual a genetic sequence which encodes for all or part of the MART-1 nucleic acid sequence is inserted into a expression vector, as described above, and introduced into the mammal to be immunized. Examples of vectors that may be used in the aforementioned vaccines include, but are not limited to, defective retroviral vectors, adenoviral vectors vaccinia viral vectors, fowl pox viral vectors, or other viral vectors (Mulligan, R. C., (1993) Science 260:926-932). The viral vectors carrying all or part of the MART-1 nucleic sequence can be introduced into a mammal either prior to any evidence of melanoma or to mediate regression of the disease in a mammal afflicted with melanoma. Examples of methods for administering the viral vector into the mammals include, but are not limited to, exposure of cells to the virus ex vivo, or injection of the retrovirus or a producer cell line of the virus into the affected tissue or intravenous administration of the virus. Alternatively the viral vector carrying all or part of the MART-1 nucleic acid sequence may be administered locally by direct injection into the melanoma lesion or topical application in a pharmaceutically acceptable carrier. The quantity of viral vector, carrying all or part of the MART-1 nucleic acid sequence, to be administered is based on the titer of virus particles. A preferred range of the immunogen to be administered may be about 106 to about 1011 virus particles per mammal, preferably a human. After immunization the efficacy of the vaccine can be assessed by production of antibodies or immune cells that recognize the antigen, as assessed by specific lytic activity or specific cytokine production or by tumor regression. One skilled in the art would know the conventional methods to assess the aforementioned parameters. If the mammal to be immunized is already afflicted with melanoma or metastatic melanoma the vaccine can be administered in conjunction with other therapeutic treatments. Examples of other therapeutic treatments includes, but are not limited to, adoptive T cell immunotherapy, coadministration of cytokines or other therapeutic drugs for melanoma.
  • Alternatively all or parts thereof of a substantially or partially purified the MART-1 protein may be administered as a vaccine in a pharmaceutically acceptable carrier. Ranges of MART-1 protein that may be administered are about 0.001 to about 100 mg per patient, preferred doses are about 0.01 to about 100 mg per patient. In a preferred embodiment, the MART-1 peptide AAGIGILTV (SEQ. ID NO. 4) (presented in single letter code) or analogs thereof is administered therapeutically or prophylactically to a mammal in need of such treatment. Alternatively, modified MART-1 peptides, examples of which are presented in Table 14 may be used. Preferred doses may be about 0.001 mg to about 100 mg, most preferred are about 0.01 mg to about 100 mg. The peptide may be synthetically or recombinantly produced. Immunization is repeated as necessary, until a sufficient titer of anti-immunogen antibody or immune cells has been obtained.
  • In yet another alternative embodiment a viral vector, such as a retroviral vector, can be introduced into mammalian cells. Examples of mammalian cells into which the retroviral vector can be introduced include, but are not limited to, primary mammalian cultures or continuous mammalian cultures, COS cells, NIH3T3, or 293 cells (ATTC #CRL 1573). The means by which the vector carrying the gene may be introduced into a cell includes, but is not limited to, microinjection, electroporation, transfection or transfection using DEAE dextran, lipofection, calcium phosphate or other procedures known to one skilled in the art (Sambrook et al. (EDS) (1989) in “Molecular Cloning. A laboratory manual”, Cold Spring Harbor Press Plainview, N.Y.). The mammalian cells expressing the MART-1 antigen can be administered to mammals and serve as a vaccine or immunogen. Examples of how the cells expressing MART-1 antigens can be administered include, but is not limited to, intravenous, intraperitoneal or intralesional. In a preferred embodiment, the part of the MART-1 nucleic acid sequence corresponding to the peptide AAGIGILTV (SEQ ID NO: 4) is inserted into the MART-1 expression vector and introduced into the mammalian cells. Alternatively, a nucleic acid sequence corresponding to MART-1 peptides which have been modified to enhance their binding to MHC molecules may be used. By way of example, the nucleic acid sequences encoding the modified peptides shown in Table 14 may be inserted into an expressions vector and introduced with mammalian cells.
  • The vaccine formulation of the present invention comprise an immunogen that induces an immune response directed against the melanoma associated antigens such as the melanoma associated MART-1 antigen. The vaccine formulations may be evaluated first in animal models, initially rodents, and in nonhuman primates and finally in humans. The safety of the immunization procedures is determined by looking for the effect of immunization on the general health of the immunized animal (weight change, fever, appetite behavior etc.) and looking for pathological changes on autopsies. After initial testing in animals, melanoma cancer patients can be tested. Conventional methods would be used to evaluate the immune response of the patient to determine the efficiency of the vaccine.
  • In yet another embodiment of this invention all, part, or parts of the MART-1 protein or MART-1 peptides or analogs thereof, or modified MART-1 peptides or analogs thereof, may be exposed to dendritic cells cultured in vitro. The cultured dendritic cells provide a means of producing T-cell dependent antigens comprised of dendritic cell modified antigen or dendritic cells pulsed with antigen, in which the antigen is processed and expressed on the antigen activated dendritic cell. The MART-1 antigen activated dendritic cells or processed dendritic cell antigens may be used as immunogens for vaccines or for the treatment of melanoma. The dendritic cells should be exposed to antigen for sufficient time to allow the antigens to be internalized and presented on the dendritic cells surface. The resulting dendritic cells or the dendritic cell process antigens can than be administered to an individual in need of therapy. Such methods are described in Steinman et al. (WO93/208185) and in Banchereau et al. (EPO Application 0563485A1) which are incorporated herein by reference.
  • In yet another embodiment of this invention T-cells isolated from individuals can be exposed to the MART-1 protein or portions thereof, or MART-1 peptides or analogs thereof or MART-1 modified peptides or analogs thereof in vitro and then administered to a patient in need of such treatment in a therapeutically effective amount. Examples of where T-lymphocytes can be isolated, include but are not limited to, peripheral blood cells lymphocytes (PBL), lymph nodes, or tumor infiltrating lymphocytes (TIL). Such lymphocytes can be isolated from the individual to be treated or from a donor by methods known in the art and cultured in vitro (Kawakami, Y. et al. (1989) J. Immunol. 142: 2453-3461). Lymphocytes are cultured in media such as RPMI or RPMI 1640 or AIM V for 1-10 weeks. Viability is assessed by trypan blue dye exclusion assay. The lymphocytes are exposed to all or part of the MART-1 protein for part or all of the culture duration. In a preferred embodiment the lymphocytes are exposed to the AAGIGILTV (SEQ ID NO: 4) peptide (presented in single letter code) at a concentration of about 1 to about 10 micrograms(ug)/ml per 107 cells for all or part of the duration of lymphocyte culture. After being sensitized to the peptide the T-lymphocytes are administered to the mammal in need of such treatment. Alternatively, the modified MART-1 peptides shown in Table 14 may be exposed to lymphocytes. Examples of how these sensitized T-cells can be administered to the mammal include but are not limited to, intravenously, intraperitoneally or intralesionally. Parameters that may be assessed to determine the efficacy of these sensitized T-lymphocytes include, but are not limited to, production of immune cells in the mammal being treated or tumor regression. Conventional methods are used to assess these parameters. Such treatment can be given in conjunction with cytokines or gene modified cells (Rosenberg, S. A. et al. (1992) Human Gene Therapy, 3: 75-90; Rosenberg, S. A. et al. (1992) Human Gene Therapy, 3: 57-73).
  • In addition to use as a vaccine, the compositions can be used to prepare antibodies to MART-1 antigen, peptides or analogs thereof, or modified MART-1 peptides and analogs thereof. The antibodies can be used directly as anti-melanoma agents. To prepare antibodies, a host animal is immunized using the MART-1 protein, peptides or analogs thereof, or modified peptides or analogs thereof as the immunogen and bound to a carrier as described above for vaccines. The host serum or plasma is collected following an appropriate time interval to provide a composition comprising antibodies reactive with the immunogen. The gamma globulin fraction or the IgG antibodies can be obtained, for example, by use of saturated ammonium sulfate or DEAE Sephadex, or other techniques known to those skilled in the art. The antibodies are substantially free of many of the adverse side effects which may be associated with other anti-cancer agents such as chemotherapy.
  • The antibody compositions can be made even more compatible with the host system by minimizing potential adverse immune system responses. This is accomplished by removing all or a portion of the Fc portion of a foreign species antibody or using an antibody of the same species as the host animal, for example, the use of antibodies from human/human hybridomas. Humanized antibodies (i.e., nonimmunogenic in a human) may be produced, for example, by replacing an immunogenic portion of an antibody with a corresponding, but nonimmunogenic portion (i.e., chimeric antibodies). Such chimeric antibodies may contain the reactive or antigen binding portion of an antibody from one species and the Fc portion of an antibody (nonimmunogenic) from a different species. Examples of chimeric antibodies, include but are not limited to, non-human mammal-human chimeras, rodent-human chimeras, murine-human and rat-human chimeras (Robinson et al., International Patent Application 184,187; Taniguchi M., European Patent Application 171,496; Morrison et al., European Patent Application 173,494; Neuberger et al., PCT Application WO 86/01533; Cabilly et al., 1987 Proc. Natl. Acad. Sci. USA 84:3439; Nishimura et al., 1987 Canc. Res. 47:999; Wood et al., 1985 Nature 314:446; Shaw et al., 1988 J. Natl. Cancer Inst. 80: 15553, all incorporated herein by reference).
  • General reviews of “humanized” chimeric antibodies are provided by Morrison S., 1985 Science 229:1202 and by Oi et al., 1986 BioTechniques 4:214.
  • Suitable “humanized” antibodies can be alternatively produced by CDR or CEA substitution (Jones et al., 1986 Nature 321:552; Verhoeyan et al., 1988 Science 239:1534; Siedleret al. 1988 J. Immunol. 141:4053, all incorporated herein by reference).
  • The antibodies or antigen binding fragments may also be produced by genetic engineering. The technology for expression of both heavy and light cain genes in E. coli is the subject the following PCT patent applications; publication number WO 901443, WO901443, and WO 9014424 and in Huse et al., 1989 Science 246:1275-1281.
  • The antibodies can also be used as a means of enhancing the immune response. The antibodies can be administered in amounts similar to those used for other therapeutic administrations of antibody. For example, pooled gamma globulin is administered at a range of about 1 mg to about 100 mg per patient. Thus, antibodies reactive with the MART-1 antigen can be passively administered alone or in conjunction with other anti-cancer therapies to a mammal afflicted-with melanoma. Examples of anti-cancer therapies include, but are not limited to, chemotherapy, radiation therapy, adoptive immunotherapy therapy with TIL.
  • Alternatively, anti MART-1 antigen antibodies can be induced by administering anti-idiotype antibodies as immunogens. Conveniently, a purified anti-MART-1 antibody preparation prepared as described above is used to induce anti-idiotype antibody in a host animal. The composition is administered to the host animal in a suitable diluent. Following administration, usually repeated administration, the host produces anti-idiotype antibody. To eliminate an immunogenic response to the Fc region, antibodies produced by the same species as the host animal can be used or the Fc region of the administered antibodies can be removed. Following induction of anti-idiotype antibody in the host animal, serum or plasma is removed to provide an antibody composition. The composition can be purified as described above for anti-MART-1 antibodies, or by affinity chromatography using anti-MART-1 antibodies bound to the affinity matrix. The anti-idiotype antibodies produced are similar in conformation to the authentic MART-1-antigen and may be used to prepare an MART-1 melanoma antigen vaccine rather than using the MART-1 protein, peptides analogs or portions thereof.
  • When used as a means of inducing anti-MART-1 antibodies in an animal, the manner of injecting the antibody is the same as for vaccination purposes, namely intramuscularly, intraperitoneally, subcutaneously, interlesionally, or the like in an effective concentration in a physiologically suitable diluent with or without adjuvant. One-or more booster injections may be desirable.
  • The MART-1 derived proteins or peptides or modified peptides of this invention are also intended for use in producing antiserum designed for pre- or post-disease prophylaxis. Here the MART-1 antigen, peptides or analogs thereof, or modified MART-1 peptides or analogs thereof is formulated with a suitable adjuvant and administered by injection to human volunteers, according to known methods for producing human antisera. Antibody response to the injected proteins is monitored, during a several-week period following immunization, by periodic serum sampling to detect the presence of anti-MART-1 serum antibodies, using an immunoassay as described herein.
  • The antiserum from immunized individuals may be administered as a prophylactic measure for individuals who are at risk of developing melanoma. The antiserum is also useful in treating an individual afflicted with melanoma for post-disease prophylaxis.
  • For both in vivo use of antibodies to MART-1 antigen and anti-idiotype antibodies and diagnostic use, it may be preferable to use monoclonal antibodies. Monoclonal anti-MART-1 antibodies or anti-idiotype antibodies can be produced as follows. The spleen or lymphocytes from an immunized animal are removed and immortalized or used to prepare hybridomas by methods known to those skilled in the art. (Goding, J. W. 1983. Monoclonal Antibodies: Principles and Practice, Pladermic Press, Inc., NY, N.Y., pp. 56-97). To produce a human-human hybridoma, a human lymphocyte donor is selected. A donor known to have a melanoma carrying the MART-1 antigen may serve as a suitable lymphocyte donor. Lymphocytes can be isolated from a peripheral blood sample or spleen cells may be used if the donor is subject to splenectomy. Epstein-Barr virus (EBV) can be used to immortalize human lymphocytes or a human fusion partner can be used to produce human-human hybridomas. Primary in vitro immunization with peptides can also be used in the generation of human monoclonal antibodies. Examples of preferred MART-1 peptides, but not limited to are, AAGIGILTV (SEQ ID NO: 4), EAAGIGILTV (SEQ ID NO: 17) and AAGIGILTVI (SEQ ID NO: 18) (peptides are presented in single letter amino acid code). Most preferably AAGIGILTV (SEQ ID NO: 4) is used as the immunogen. Alternatively, peptides derived from the MART-1 amino acid sequence and modified to enhance binding of the peptide to a MHC Class I molecule may also be used. By way of example the modified peptides shown in Table 14 may be used as the immunogen.
  • Antibodies secreted by the immortalized cells are screened to determine the clones that secrete antibodies of the desired specificity. For monoclonal MART-1 antigen or peptide antibodies, the antibodies must bind to MART-1 antigen or peptide. For monoclonal anti-idiotype antibodies, the antibodies must bind to anti-MART-1 antibodies. Cells producing antibodies of the desired specificity are selected.
  • The antibodies or chimeric antibodies described herein may also be coupled to toxin molecules radio-isotopes and drugs by conventional methods (Vitetta et al. (1991) in “Biologic Therapy of Cancer” De Vita V T, Hellman S., Rosenberg, S. A. (eds) J.B. Lippincott Co. Philadelphia; Larson, S. M. et al. (1991) in “Biological Therapy of Cancer” De Vita V. T., Hellman S., Rosenberg, S. A. (eds) J.B. Lippincott Co., Philadelphia). Examples of toxins to which the antibodies may be coupled to include, but are not limited to, ricin or diphtheria toxin. Examples of drugs or chemotherapeutic agents include, but are not limited to, cyclophosphamide or doxorubcin. Examples of radioisotopes, include, but are not limited to, 131I, Antibodies covalently conjugated to the aforementioned agents can be used in cancer immunotherapy for treating melanoma.
  • Local administration to the afflicted site may be accomplished through means known in the art, including, but not limited to, topical application, injection, and implantation of a porous device containing cells recombinantly expressing the infusion, implantation of a porous device in which the MART-1 antibodies or chimeric antibodies, antibodies coupled to toxins, drugs or radiolabels or portions thereof are contained.
  • The above described antibodies and antigen binding fragments thereof may be supplied in kit form alone, or as a pharmaceutical composition for in vivo use. The antibodies may be used for therapeutic uses, diagnostic use in immunoassays or as an immunoaffinity agent to purify the MART-1 protein or peptides as described herein.
  • The present invention also provides a substantially purified and isolated nucleic acid sequence, designated c(complementary)DNA25 (FIG. 4; SEQ. ID. NO. 26) which encodes a second melanoma recognized by tumor infiltrating lymphocytes. The TIL which recognize the melanoma antigen encoded by cDNA25 are associated with in vivo tumor rejection. The TIL recognized the melanoma antigen encoded by cDNA25 in the context of HLA-A2. Comparison of the cDNA25 nucleic acid sequence (FIG. 4; SEQ. ID NO 26) with the nucleic acid sequences for genes encoding a melanocyte-melanoma specific protein gp100 shows this sequence to be similar, but distinct, from the previously identified sequences for gp100. Previously identified sequences for gp100 include gp100 (GenBank Accession No. M32295; also designated gp95), Pmel 17 (GenBank Accession No. M77348; Kwon et al., (1991) Proc. Natl. Acad. Sciences (USA) 88:9228-9232) and ME20 (Maresh et al. (1994) DNA and Cell Biology 13:87-95).
  • The cDNA25 sequence provided herein (FIG. 4; SEQ. ID. NO. 26) differs from the previously reported gp100 sequence in Genbank (Genbank Accession No. M32295) by two nucleotides, from the Pmel 17 sequence (Kwon et al. (1991) Proc. Natl. Acad. Sciences (USA) 88: 9228-9232) by three nucleotides and a twenty one base pair deletion, and from the ME20 sequence (Maresh et al. (1994) DNA and Cell Biology 13:87-95) by a single nucleotide difference. At the amino acid level, the protein encoded by cDNA25 differs from the gp100 in GenBank (GenBank Accession #M32295) by one amino acid at position 162, by a two amino acid difference at positions 162 and 274, compared to Pmel 17 and did not contain 7 amino acids that existed in Pmel 17 at positions 588-594. Therefore, cDNA25 appears to encode for a different form of the gene for gp100. The differences between the cDNA25 nucleic acid sequence (FIG. 4; SEQ. ID. NO. 26) and amino acid sequence (FIG. 5A; SEQ. ID. NO. 27) and previously reported gp100 sequences may be due to polymorphisms, allelic variations, or to mutations within the tumor. Experiments with mouse tumors have shown that new antigens recognized by T-cells can result from point mutation in the coding region of the inactive gene (Boon, T (1992) Advances in Cancer Research 58:177-210).
  • This invention also provides immunogenic peptides derived from the gp100 protein sequence provided herein. (FIG. 5A; SEQ. ID. NO. 27). These immunogenic peptides represent the antigenic portions of the gp100 protein (FIG. 5A; SEQ. ID. NO. 27) recognized by TIL. Examples of immunogenic peptides include, but are not limited to, LLDGTATLRL (peptide G10-4; SEQ ID NO: 33; FIG. 4, nucleotides 1407-1436), VLYRYGSFSV (peptide G10-5; SEQ ID NO: 34; FIG. 4 nucleotides 1464-1493), ALDGGNKHFL (peptide G10-22; SEQ ID NO: 35; FIG. 4, nucleotides 708-737), VLKRCLLHL (peptide G9-19 SEQ ID NO: 36; FIG. 4, nucleotides 48-74), VLPSPACQLV (peptide G10-8; SEQ ID NO: 37; FIG. 4 nucleotides 1668 to 1797) SLADTNSLAV (peptide G10-9; SEQ ID NO: 38; FIG. 4 nucleic acids 1746 to 1775) SVSVSQLRA (peptide G9-216; SEQ ID NO:39; FIG. 4, nucleotides 684 to 710) YLEPGPVTA (peptide G9-280; SEQ ID NO:40; FIG. 4; nucleotides 876-902) or LNVSLADTN (peptide G10-400; SEQ ID NO:41; FIG. 4; nucleotides 1736-1764) KTWGQYWQV (peptide G9154; SEQ ID NO:46; FIG. 5A; amino acids 154 to 162), KTWGQYWQVL (peptide G10154 SEQ ID NO:47; FIG. 5A; amino acids 154 to 163), ITDQVPFSV (peptide G9209, SEQ ID NO:48; FIG. 5A; amino acids 209 to 217) and TITDQVPFSV (peptide G10208; SEQ ID NO: 49; FIG. 5A; amino acids 208 to 217). This invention further includes analogs of these immunogenic peptides derived from the gp100 amino acid sequence (FIG. 5A; SEQ ID NO: 27). The term analog includes any peptide which displays the functional aspects of these immunogenic peptides. The term analog also includes conservative substitution or chemical derivative of the peptides as described above. These immunogenic peptides may be synthetically or recombinantly produced in the same manner or fashion as described above for MART-1.
  • In yet another embodiment of this invention, immunogenic peptides derived from the gp100 sequence (FIG. 5A; SEQ ID NO: 27, are modified to increase immunogenicity by enhancing the binding of the peptide to MHC molecule with which the peptide is associated when presented to T-cells. By way of example, modifications may include the substitution, deletion or addition, of one or more amino acids within the immunogenic peptide sequence, or insertion of amino acids within the given immunogenic peptide sequence or derivitization of existing amino acids within the given immunogenic peptide sequence or mutation of the amino acids within the given immunogenic peptide sequence. In a preferred modification at least one amino acid is substituted or replaced in the given immunogenic peptide sequence. Any amino acid composing the given immunogenic peptide sequence may be modified in accordance with this invention. Any amino acid may be used to substitute or replace a given amino acid within the immunogenic peptide sequence. Modification may occur at any amino acid position within the immunogenic gp100 peptide. Modified gp100 peptides is intended to include any modified immunogenic gp100 peptide exhibiting enhanced binding with the MHC molecule with which it is associated when presented to the T-cell.
  • By way of example peptides recognized by T cells in the context of HLA-A2 are 9 to 10 amino acids in length. Preferably for enhanced binding of the peptide to HLA-A2 the second position and last position in the peptide are hydrophobic amino acids preferably aliphatic hydrophobic amino acids. The second position may be any aliphatic hydrophobic amino acid such as, but not limited to, leucine, methionine, isoleucine, valine, threonine, glycine or alanine. The last position of the peptide ( position 9 or 10 depending on the peptide's length) may be any aliphatic hydrophobic amino acid, such as but not limited to valine, leucine, alanine, leucine, isoleucine, glycine, methionine, valine, or threonine.
  • The first and third positions of the immunogenic peptide may be substituted or replaced with any amino acid, preferably, hydrophobic aliphatic amino acids, or aromatic amino acids. Examples of amino acids that may be used at the first or third position of the peptide include, but are not limited to, alanine, leucine, lysine, isoleucine, glycine, methionine, valine, threonine, tryptophan, phenylalanine, serine, lysine or tyrosine.
  • Examples of gp100 peptides that may be modified in accordance with the present embodiment include, but is not limited to LLDGTATLRL (peptide G10-4; SEQ ID NO: 33; FIG. 4, nucleotides 1407-1436), VLYRYGSFSV (peptide G10-5; SEQ ID NO: 34; FIG. 4 nucleotides 1464-1493), ALDGGNKHFL (peptide G10-22; SEQ ID NO: 35; FIG. 4, nucleotides 708-737), VLKRCLLHL (peptide G9-19 SEQ ID NO: 36; FIG. 4, nucleotides 48-74), VLPSPACQLV (peptide G10-8; SEQ ID NO: 37; FIG. 4 nucleotides 1668 to 1797) SLADTNSLAV (peptide G10-9; SEQ ID NO: 38; FIG. 4 nucleic acids 1746 to 1775) SVSVSQLRA (peptide G10-216; SEQ ID NO:39; FIG. 4, nucleotides 684 to 710) YLEPGPVTA (peptide G9-280; SEQ ID NO:40; FIG. 4; nucleotides 876-902) or LNVSLADTN (peptide G10-400; SEQ ID NO:41; FIG. 4; nucleotides 1736-1764) KTWGQYWQV (peptide G9154; SEQ ID NO:46; FIG. 5A; amino acids 154 to 162), KTWGQYWQVL (peptide G10154 SEQ ID NO:47; FIG. 5A; amino acids 154 to 163), ITDQVPFSV (peptide G9209, SEQ ID NO:48; FIG. 5A; amino acids 209 to 217) and TITDQVPFSV (peptide G10208; SEQ ID NO: 49; FIG. 5A; amino acids 208 to 217).
  • By way of example modified gp100 peptides derived from the immunogenic gp100 peptide KTWGQYWQV (SEQ ID NO:46) may have the formula X1X2X3GQYWQX4 wherein:
  • X1 may be any amino acid, preferably any hydrophobic aliphatic amino acid, or aromatic amino acid. Examples of amino acids that may be used include, but are not limited to, alanine, leucine, lysine, isoleucine, glycine, methionine, valine, threonine, tryptophan, phenylalanine, lysine or serine, aspartic acid or tyrosine;
  • X2 may be any hydrophobic amino acid, preferably any aliphatic hydrophobic amino acid. Examples of amino acids that may be used include, but are not limited to, leucine, methionine, isoleucine, alanine, threonine, glycine, or valine. Most preferably leucine, methionine or isoleucine.
  • X3 may be any amino acid, preferably any hydrophobic aliphatic amino acid or aromatic amino acid. Examples of amino acids that may be used include, but are not limited to, alanine, leucine, lysine, isoleucine, glycine, methianine, valine, threonine, tryptophan, phenylalanine, serine, lysine, or tyrosine;
  • X4 is any hydrophobic amino acid, preferably an aliphatic hydrophobic amino acid. Examples of amino acids that may be used include, but are not limited to, valine, leucine, isoleucine, methionine, alanine, threonine, or glycine.
  • Examples of modified peptides are shown in Table 15. A preferred modified peptide is KIWGQYWQV (G9-154-2I).
  • Alternatively, the immunogenic gp100 ITDQVPFSV (G9-209; SEQ ID NO:48) may be modified, such modified peptides may have the general formula X1X2X3QVPFSX4 wherein:
  • X1 may be any amino acid, preferably any hydrophobic aliphatic amino acid, or aromatic amino acid. Examples of amino acids that may be used include, but are not limited to, leucine, methionine, alanine, isoleucine, valine, threanine, glycine, lysine, phenylalanine, tryptophan, or tyrosine, aspartic acid or serine;
  • X2 is any hydrophobic amino acid, preferably a hydrophobic aliphatic amino acid. Examples of amino acids that may be used include, but are not limited to, leucine, methionine, alanine, isoleucine, valine, threanine, or glycine;
  • X3 may be any amino acid, preferably any hydrophobic, aliphatic amino acid or aromatic amino acid. Examples of amino acids that may be used include, but are not limited to, leucine, methionine, alanine, isoleucine, valine, threonine, glycine, lysine, phenylalanine, tryptophan, tyrosine, aspartic acid or serine;
  • X4 may be any hydrophobic amino acid, preferably any hydrophobic aliphatic amino acid. Examples of amino acids that may be used include, but are not limited to, leucine, methionine, alanine, isoleucine, valine, or threonine;
  • Examples of modified peptides derived from ITDQVPFSV are shown in Table 16. Preferably the peptide FLDQVPFSV (peptide G9-209-1F2L) is used.
  • By way of example modified gp100 peptides derived from the immunogenic gp100 peptide YLEPGPVTA (G9-280; SEQ ID NO:40) may also be modified to enhance binding to MHC Class I molecules, preferably HLA-A2 or subtypes thereof.
  • The modified peptides may have the general formula X1X2X3PGPVTX4 wherein:
  • X1 may be any amino acid, preferably a hydrophobic aliphatic amino acid or aromatic amino acid. Examples of amino acids that may be used include, but are not limited to, leucine, methionine, alanine, isoleucine, valine, threanine, glycine, lysine, phenylalanine, tryptophan, or tyrosine, aspartic acid or serine;
      • X2 may be any hydrophobic amino acid, preferably an aliphatic hydrophobic amino acid. Examples of amino acids that may be used include, but are not limited to leucine, methionine, alanine, isoleucine, valine, threonine, or glycine;
  • X3 may be any amino acid, preferably any hydrophobic aliphatic amino acid, or aromatic amino acid. Examples of amino acids that may be used include, but are not limited to, leucine, methionine, alanine, isoleucine, valine, threanine, glycine, lysine, phenylalanine, tryptophan, tyrosine, aspartic acid or serine;
  • X4 may be any hydrophobic amino acid preferably an aliphatic hydrophobic amino acid. Examples of amino acids that may be used include but are not limited to, leucine, methionine, alanine, isoleucine, valine, threonine, or glycine.
  • Examples of modified peptides derived from YLEPGPVTA (G9-280; SEQ ID NO:40) are shown in Table 17. A preferred modified peptide is YLEPGPVT (G9-280-9V).
  • This invention further includes analogs of these modified peptides derived from the gp-100 sequence (FIG. 5A; SEQ ID NO:27). The term analog is intended to include any peptide which displays the functional aspects of these modified peptides as described above. These modified peptides may be synthetically or recombinantly provided by conventional methods.
  • In another embodiment the immunogenic peptides (SEQ ID NO: 33 to 38 and 46 to 49) derived from the gp100 amino acid sequence (SEQ ID NO: 27) or modified gp100 peptides as shown in Tables 15-17 or analogs thereof, may be used as a vaccine either therapeutically or prophylactically. When provided, prophylactically the vaccine is provided in advance of any evidence of melanoma. The prophylactic administration of these peptides should serve to prevent or attenuate melanoma in a mammal.
  • In a preferred embodiment, mammals, preferably humans, at high risk for melanoma are prophylactically treated with these vaccines. Alternatively, the vaccine may be provided therapeutically to enhance the patients own immune response to the tumor antigen prescribed on the melanoma or metastatic melanoma. The vaccine, which acts as an immunogen, may be a cell, cell lysate from cells transfected with a recombinant expression vector carrying a nucleic acid sequences encoding gp100 immunogenic peptide or a culture supernatant containing the expressed protein. Expression vectors into which nucleic acid sequences encoding these immunogenic peptides may be introduced are the same as those described above for MART-1. Alternatively, the immunogen is a partially or substantially purified recombinant gp100 peptide or analog thereof.
  • While it is possible for the immunogen to be administered in a pure or substantially pure form, it is preferable to present it as pharmaceutical compositions, formulations or preparations as described above for MART-1. Vaccination can be conducted by conventional methods previously described above for MART-1.
  • The gp100 immunogenic peptides and nucleic acids sequences encoding them may be used in bioassays, or to generate antibodies in the same manner or fashion as described above for MART-1.
  • In yet another embodiment of this invention, multivalent vaccines against one or more melanoma antigens are provided. Such multivalent vaccines may comprise all or part of the MART-1 protein peptides or modified peptides or gp100 peptides or modified peptides or combinations thereof.
  • Previous identification of genes encoding melanoma antigens have utilized PBL isolated from melanoma patients immunized or pretreated with antigens (Van Der Bruggen et al. (1991) Science 254: 1643-1647; Brichard et al. (1993) J. Exp. Med. 178: 489-495; Traversari, C., et al. (1992) J. Exp. Med. 176: 1453-1457). A preferred strategy is to identify genes coding for tumor antigens that are recognized by TIL from tumor-bearing patients, in the absence of immunization of said patients. Such a strategy enhances the possibility that the genes identified code for antigens involved in the natural immune response against the growing cancer. Thus, this invention also provides a method of identifying genes encoding melanoma antigens utilizing cDNA expression cloning using tumors infiltrating lymphocytes isolated (TIL) from the tumor of patients afflicted with melanoma. The method comprises the following steps: (a) isolating tumor infiltrating lymphocytes from the tumor of a mammal afflicted with melanoma; (b) introducing a melanoma cDNA library into a mammalian cell line; (c) exposing said mammalian cells to said TIL; (d) screening for expression of an antigen encoded by said cDNA in said mammalian cells recognized by said TIL; and (e) isolating said cDNA corresponding to said antigen. The tumor infiltrating lymphocytes in step (a) may be isolated from patients afflicted with melanoma including, but not limited to, the melanoma lesion, subcutaneous tissue or visceral organs. Examples of cells that may be used to prepare the cDNA library used in step (b), include, but are not limited to, fresh or cultured melanoma cells. Preferably, the cDNA library is introduced into mammalian cells not expressing melanoma antigens. If non human mammalian cells or human cells not expressing the desired HLA haplotype for recognition by the TIL are used in step (b), such cells can be cotransfected with an HLA gene as described below. Examples of cells which can be used in step (b), include but are not limited to, tumor cell lines, such as breast cancer cell line MDA 231 (ATCC #HTB26), or COS 7 cells (ATCC #CRL 1651). Examples of MHC genes which can be used include, but are not limited to, HLA-A, HLA-B, and HLA-C genes, preferably HLA-A2 and subtypes thereof (Zemmour, J. et al. (1992) Tissue Antigens 40:221-228). The appropriate MHC gene to be used is determined by the haplotype of the tumor cells which were the source for the cDNA library. Standard methods can be used to determine the haplotype recognized by the TIL isolated (ASHI Laboratory Manual (2nd Edition 1990). Examples of how to evaluate recognition of the cells containing the cDNA clone expressing an antigen recognized by the TIL includes, but is not limited to, γ-interferon assays, TNF secretion (Van de Bruggen et al., (1991) Science 254:1643-1647) or lysis of cells transfected with cDNA encoding for the recognized antigen. Such assays are performed by conventional methods known to one skilled in the art. Melanoma antigens can be isolated by or rescued by PCR using primer specific to flanking site of vector containing the cDNA. Examples of how to isolate the cDNA corresponding to the antigen recognized by the TIL include, but are not limited to, PCR.
  • Once the genes or nucleic acid sequences encoding melanoma antigens are identified, the next step is to determine the antigenic portion or epitope of the protein encoded by these genes. Therefore, in yet another embodiment of this invention, a method is provided for assessing the immunogenicity of peptides derived from the predicted amino acid sequences of the MART-1 protein (FIG. 1; SEQ ID NO: 2) or a gp100 protein (FIG. 5A; SEQ ID NO: 27). The method comprises the steps of: (a) preparing a plurality of peptides based on the MART-1 (FIG. 1; SEQ ID NO: 2) or gp100 (FIG. 5A; SEQ ID NO: 27) amino acid sequence; (b) incubating at least one of said peptides with a mammalian cell line; (c) exposing said mammalian cells incubated with said peptide to tumor infiltrating lymphocytes (TIL); and (d) screening for recognition of TIL with said cells incubated with said peptide. It is preferred that peptides of about 25 to 5 amino acids be used, more preferably 20 to 10 amino acids and most preferably 9-10 amino acids. Examples of cells that may be used in step (b) include, but are not limited to, T2-cells, (Cerundolo, V. et al. (1990) Nature, 345: 449-452) or EBV transformed B cell lines (Topalian et al. (1989) J. Immunol. 142: 3714-3725). Examples of how to assess recognition of the cells incubated with peptide include but is not limited to, 51CR release cytotoxicity assay (Cerundolo, V. et al. (1990) Nature 345:449-452.) or lymphokine assays such as γ-IFN or TNF secretion. (Schwartzentruber, D. et al., (1991) J. of Immunology 146:3674-3681).
  • T cells recognize antigen complexed with MHC Class 1 molecules. The MHC locus in all mammalian species contains numerous genes and is highly polymorphic. Different MHC molecules or haplotypes types bind different antigens. In humans the HLA complex contains the HLA-A, HLA-B and HLA-C gene loci which encode class I molecules. Lymphocytes will recognize tumor antigens on the context of HLA Class 1 molecule. If the cells containing the recombinant MART-1 expression vector are to be screened by the TIL but are not human cells, such as COS cells, or do not express a desired haplotype an expression vector containing an MHC Class 1 gene may also be introduced into the cells. (See Example 1) This, represents yet another alternative embodiment of the invention. Cells expressing MART-1 antigens and HLA antigens can by screened with TIL to detect the presence of tumor antigens in the context of a specific MHC Class 1 restriction type. The appropriate haplotype is determined by the haplotype of the tumor from which the library is derived. Examples of MHC Class I genes that may be used include, but are not limited to, HLA-A, HLA-B and HLA-C genes. Examples of preferred MHC specificities or restriction types include, but is not limited to HLA-A1, HLA-A2, such as the HLA-A2.1 subtype, or HLA-A24 (Zemmour, J. et al. (1992) Tissue Antigens 40:221-228). Most preferred is the HLA-A2.1 gene.
  • Veterinary uses are also intended to be encompassed bit the compositions and therapeutic applications described herein.
  • All books, articles, and patents referenced herein are incorporated by reference in their entirety. The following examples illustrate various aspects of the invention and in no way intended to limit the scope thereof.
  • Cloning of a Gene Coding for a Shared Human Melanoma Antigen Recognized by Autologous T Cells Infiltrating into Tumors EXAMPLE 1 Generation of Cytotoxic T Lymphocytes (CTL) and Culture of Cell Lines
  • −54 CTL were generated from excised tumor specimens by culturing a suspension of cells with 6000 IU/ml of IL-2 (Cetus-Oncology Division, Chiron Corp. Emeryville, Calif.) for 30-70 days as described in Kawakami, Y., et al. (1988) J. Exp. Med. 168:2183-2191. TIL501 and TIL1235 were predominantly CD8+ and were derived from the tumor specimens of patients with advanced metastatic melanoma. The CD8+ T cell clone, TIL501.A42, was established by limiting dilution methods and cultured with 120 IU/ml of IL-2 plus irradiated (once a week for 4 to 6 times) autologous tumor cells.
  • Melanoma-cell lines, 397mel, 501mel, 526mel, 537mel, 624mel, 888mel, 952mel, and Epstein-Barr virus (EBV) transformed B cell lines, 501EBVB, 836EBVB were established in our laboratory and cultured in RPMI1640 (GIBCO/Lifetechnologies, Grand Island N.Y.) medium containing 10% fetal calf serum (FCS) (Biofluids, Rockville Md.). (Topalian et al., (1989) J. Immunol. 142: 3714-3725) Normal cultured melanocytes, NHEM483, NHEM493, NHEM527, NHEM529, NHEM530, NHEM533, NHEM616 and NHEM680 were purchased from Clonetics, San Diego, Calif., FM725, FM801, FM902 were provided by M. Herlyn, Wistar Institute, Philadelphia Pa., HA002 was provided by R. Halaban, Yale university, New Haven, Conn. and cultured in melanocyte growth medium (MGM, Clonetics). Melanoma cell lines, C32, RPMI7951, WM115, A375, HS695T, Malme3M, colon cancer cell lines, Collo, SW480, WiDr, breast cancer cell lines, MDA231, MCF7, HS578, ZR75, neuroblastoma cell line, SK—N—SH, glioma cell lines, U138MG, HS683, H4, sarcoma cell line 143B, embryonal kidney cell line 293 transformed with adenovirus type 5 were purchased from ATCC, Rockville, Md. Renal cancer cell lines, UBK108 and UOK117 were provided M. Linehan NIH, Bethesda, Md. The small cell lung cancer cell line, H1092 was provided by J. D. Minna, University Texas Southwestern, Dallas, Tex. Ewing's sarcoma cell lines, TC71, RD-ES, 6647 were provided by M. Tsokos, NIH, Bethesda, Md. The neuroblastoma cell line, SK—N-AS was provided by O. M. El Badry, NIH, Bethesda, Md. The plasmacytoma cell line, HMY-C1R and the M1 fibroblast cell line were provided by W. Biddison, NIH, Bethesda, Md. Kidney epithelial cells, KAM, WLC were provided by D. J. Hazen-Martin and D. A. Sens, Medical University of South Carolina, Charleston, S.C. The monkey kidney cell line, COS7, was provided by W. Leonard, NIH, Bethesda, Md.
  • Cytotoxicity Assay
  • 51Cr release assays were performed as described in Kawakami, Y., et al., (1988) J. Exp. Med. 168:2183-2191. Briefly, 5000 target cells labeled with 51Cr were mixed with varying numbers of effector cells and incubated for 5 hours (h). Then supernatants were collected, radioactivity was measured and the percent specific lysis was calculated.
  • IFN-γ Release Assay
  • Fifty to one hundred thousand responder cells and 4×104-105 stimulator cells were mixed in 300 ul of AIM-V medium containing 120 IU/ml IL-2 per well in a 96 flat-well microplate. After incubation for 20 h, 100 ul of supernatants were collected and added to an enzyme-linked immunosorbent assay (ELISA) plate (Immunoplate MaxiSorp, Nunc, Denmark) coated with anti-human IFN-γ monoclonal antibody (mAb) (Biosource, Camerillo, Calif.). After overnight incubation at 4° C., the plates were washed 3 times and 100 ul of a 1:2000 dilution of rabbit anti-human IFN-γ polyclonal antibody (Ab)(Biosource, Camerillo, Calif.) was added and incubated at 37° C. for 2 h. Plates were washed 3 times, and 100 ul of a 1:2000 dilution of alkaline phosphatase labelled goat anti-rabbit IgG polyclonal antibody (Ab) (Boehringer Mannheim, Indianapolis, Ind.) was added. After a 1 h incubation at 37° C., 100 ul of 4 mg/ml p-Nitrophenyl phosphate (Sigma, St Louis, Mo.) was added, incubated for 10-20 min at room temperature in the dark, and 25 ul of 1N NaOH was added to stop the reaction. Optical density was measured at 405 nm wave length and the concentration of IFN-γ was calculated in comparison to recombinant IFN-γ standards (Biogen, Cambridge, Mass.) measured in the same assay.
  • cDNA Expression Cloning
  • A cDNA library was constructed from poly A RNA from the HLA-A2+ melanoma cell line, 501mel as described in (Miki, T., et al., (1989) Gene; 83:137-146 Miki et al. (1991) Proc. Nat. Acad. Sci. USA 88:5167-5171). Briefly, first strand cDNA was synthesized with a linker primer GGACAGGCCGAGGCGGCC(T)40 followed by second strand cDNA synthesis. After treatment with T4 DNA ligase, an SfiI adaptor consisting of two oligonucleotides, CCAATCGCGACC and GGTCGCGATTGGTAA was ligated to the end of the cDNA. The cDNA was digested with SfiI and the digested fragment was isolated by passing through a spun column. The cDNA was then mixed with bacteriophage λpCEV27 (Miki, T. et al., (1991) Proc. Natl. Acad. Science (USA) 88: 5167-5771) vector arms prepared by SfiI digestion and in vitro packaging was performed.
  • To screen for melanoma antigens, 10 ug of the amplified cDNA library containing approximately 107 clones was transfected into the HLA-A2+ antigen non-expressing cell lines, MDA231 clone 7 and A375 clone 1-4 using a modified calcium-phosphate method (Mammalian Transfection Kit, Stratagene). After G418 (BRL, Gaithersburg, Md.) selection, individual colonies were isolated and cultured in 96 well microplates and replica plates were made. A mixture of 5×104 TIL1200 and 5×104 TTL1235 was added to the wells of the microplates containing the growing transfectants that were near confluence and incubated for 20 h. Supernatants were collected and IFN-γ was measured by ELISA.
  • Polymerase chain reaction(PCR) was performed to rescue the transfected genes from the genomic DNA of positive transfectants using SP6 and T7 primers which flank the inserted genes. The amplified products were cloned in the pCRII vector (Invitrogen, San Diego, Calif.). For cDNA clones 22 and 23, a Hind III and XhoI fragment containing the full length cDNA was subcloned into the expression vector pcDNA3 (Invitrogen, San Diego, Calif.).
  • To test whether the cloned cDNAs encode tumor antigens, the pcDNA3 containing the cloned genes were transiently transfected into the COS7 cell line by the DEAE dextran method (Seed, B. and Aruffo, A. (1987) Proc. Natl. Acad. Sci. (USA) 84:3365-3369). Briefly, 3×105 cells per well in 6 well plates were incubated at 37° C. for 4 hours (h) in 0.75 ml DMEM containing 10 ug of DEAE dextran (Sigma), 0.1 mM chloroquine and lug of the pcDNA3 containing the cloned genes and/or the pcDNA-HLA-A2.1 (Zemmour, J. et al. (1992) Tissue Antigens 40: 221-228). After medium was removed, 10% DMSO solution in HBSS buffer was added and incubated for 2 min. The cells were washed once with PBS and incubated in 7.5% FCS DMEM for 2 days. The 293 cell line was transiently transfected using lipofectamine (BRL, Gaithersburg, Md.) according to the manufacturer's recommendation. After incubation, the ability of the transfected COS7 or 293 cells to mediate IFN-γ release from TIL was assessed. The expression of the HLA-A2 gene was tested by flow cytometry. Stable transfectants were made by the calcium-phosphate method and individual colonies and pooled transfectants were tested for reactivity to TIL by cytotoxicity and IFN-γ release assays.
  • DNA sequencing of the cloned genes was performed by dideoxy chain termination method with dGTP and 7-deaza-dGTP. DNA and protein sequences were analyzed by the GCG program with GeneBank, and EMBL Data Library nucleotide databases and SWISS-PROT, PIR, GenPept, Brookhaven Protein Data Bank protein databases.
  • Northern Blot Analysis
  • Total RNA was isolated by the guanidine-isothiocyanate-cesium chloride centrifugation method. (Chirgwin, J. M. et al. (1979) Biochemistry 18: 5294). Total RNA from normal tissue was purchased from Clontech, (Palo Alto, Calif.). Ten to twenty micrograms of total RNA was subjected to electrophoresis in a 1% agarose formaldehyde gel and transferred to a nylon membrane (Duralon-UV membranes, Stratagene, La Jolla, Calif.) The Sal I digested fragment containing the full length cDNA from clone 22 and the β-actin cDNA (Clontech) were labeled by random priming and used as a probe. Hybridization with the probe was performed according to the QuikHyb protocol (Stratagene) at 68 C for 2-16 h. Membranes were washed two times with 2×SSC/0.1% SDS at 60 C for 15 minutes (min) and once with 0.1×SSC at 60 C at for 30 min and then autoradiography was performed.
  • Characterization of Cultured TIL from Melanoma Patients
  • Multiple TIL lines were established from HLA-A2+ melanoma patients and tested for lysis of melanoma cell lines from HLA-A2+ and HLA-A2 patients. HLA typing of patients is performed by conventional HLA typing techniques. HLA-A2 was selected because it is the most frequently expressed class I MHC antigen (about 50% of individuals) and has been shown to be a dominant restriction element for the recognition of melanoma antigens (Crowley, N. J., et al. (1991), J. Immunol. 146:1692-1694). TIL501, TIL1235 and TIL1200 exhibited specific recognition of shared melanoma antigens in an HLA-A2 restricted fashion. TIL501.A42 was a T cell clone established from TIL501 by limiting dilution. These TIL caused lysis or released cytokines including IFN-γ, TNFα and GM-CSF when cocultured with a variety of HLA-A2+ melanoma or melanocyte cell lines but not HLA-A2 melanoma lines or HLA-A2+ non-melanoma cell lines including the breast cancer cell line, MDA 231. Two representative experiments are shown in Table 1. Thus, these CTL seemed to recognize a non-mutated peptide derived from a melanocyte lineage specific antigen.
  • Cloning of cDNA Coding for Melanoma Antigens Recognized by T Cells
  • A cDNA library from the HLA-A2+ 501mel melanoma cell line was transfected into two highly transfectable HLA-A2.1+ cancer cell lines, MDA231 and A375. These cell lines were not lysed by melanoma specific TIL but were lysed by HLA-A2 restricted influenza M1 specific CTL after incubation with the M158-66 peptide (GILGFVFTL; single letter code) derived the influenze matrix protein or infection with a recombinant vaccinia virus containing the M1 gene (data not shown). Thus, these cell lines exhibited normal antigen processing and presenting ability but were not lysed by these melanoma-specific TIL because of the lack of expression of the relevant melanoma antigens. After selection with G418, approximately 6700 transfected clones from each cell line were isolated and grown in microplates. Using the IFN-γ release assay, 21 MDA231 and 27 A375 positive clones were isolated and rescreened. Of these clones, eight MDA231 and seven A375 clones were positive in a second screening assay.
  • In order to rescue the integrated genes, PCR using genomic DNA from these positive transfectants was performed with SP6 and T7 primers flanking the insert genes. Eight genes that were amplified from the seven transfectants which showed 1 to 2 sharp bands, including a 1.6 Kb band from MDA-22 and MDA-23 transfectants, were subcloned into the pCRII cloning vector, and then further cloned into the pcDNA3 eukaryotic expression vector. The 1.6 Kb band detected by Northern blot analysis with the cDNA 22 probe suggested that this fragment was a full length cDNA.
  • Transient transfection of the expression vector pcDNA3 containing the cDNA from clones 22 or 23 into either COS7 or 293 cells along with the HLA-A2.1 gene conferred reactivity to TIL1235 and TIL501.A42 as demonstrated by the specific release of IFN-γ (Table 2, Experiments 1 and 2). Stable transfection of these cDNA fragments into MDA 231 or A375mel cell lines also conferred reactivity to TIL1235 and TIL501.42 (Table 2, Experiment 3). TIL501.A42 could lyse MDA231 stably transfected with cDNA 22 (data not shown). These results indicated that these cDNAs encode a melanoma antigen recognized by HLA-A2 restricted TIL from melanoma patients. Transfection of another clone, MDA-25 stimulated the release of interferon-γ only from TIL 1200.
  • TABLE 1
    Specificity of TIL 501.A42 and TIL 1235 for Melanomas and Melanocytes
    A. Lytic Specificity* B. Cytokine Release Specificity**
    TIL1235 Stimulator** TIL1235 TIL586++
    Target* HLA-A2 TIL501.A42 % Lysis LAK+ Cells HLA-A2 TIL501.A42 pg IFN- γ/ml
    501mel + 54 51 78 501mel + 647 219 <50
    526mel + 25 33 74 586mel <50 <50 1034
    624mel + 23 27 75 NHEM493 + 1835 850 <50
    952mel + 10 11 75 NHEM527 + 1638 749 <50
    Malme3M + 36 41 70 NHEM530 + 1224 2532 <50
    C32 + 17 23 82 NHEM533 + 300 251 <50
    RPMI7951 + 1 6 67 NHEM616 + 635 423 <50
    WM115 + −2 3 68 FM725 + 5975 1471 <50
    HS695T + 1 2 87 FM801 + 1375 893 62
    397mel −1 0 70 NHEM483 <50 <50 <50
    MDA231 + 0 3 94 NHEM680 <50 <50 548
    HA002 <50 <50 <50
    TIL 501.A42 and TIL1235 lysed most HLA-A2 melanoma cell lines and secreted IFN-γ when cultured with HLA-A2 melanomas and melanocytes.
    *51Cr release assay was performed at E:T = 20:1 for TIL501.A42, at 40:1 for TIL1235. All targets were melanoma cell lines except for MDA231 which was a breast cancer cell line.
    **IFN-γ in the supernatant was measured after TIL and stimulator cells were coincubated for 20 hr. 501mel and 586mel are melanoma cell lines. All others were normal melanocyte cell lines.
    +LAK: Lymphokine activated killer cells.
    ++TIL586 is class I MHC restricted melanoma specific TIL, not restricted by HLA-A2.
  • Characterization of this cDNA revealed it to be similar, but distinct, from a previously described melanoma antigen gp100 recognized by monoclonal antibody HMB45. This clone is described in more detail in Example 3.
  • The cDNA sequence of clones 22 and 23 were identical except at a single base that was believed to be a change introduced by PCR. Two other independently amplified fragments were also sequenced to clarify this region and the consensus sequence is shown in FIG. 1. The longest open reading frame in this gene consists of 354 bases corresponding to a 118 amino acid protein of 13 kd. This sequence did not show significant similarity to any complete nucleotide or protein sequences in established databases. Amino acids 27-47 consist of a hydrophobic region that may contain the HLA-A2 binding peptides (Falk, K., et al. (1991), Nature 351:290-296; Hunt, D. F., et al. (1992), Science 255:1261-1263; Ruppert, J., et al. (1993), Cell 74:929-937; Nijman, H. W., et al. (1993), Eur. J. Immunol. 23:1215-1219). The antigen encoded by the cDNA 22 and 23, was designated the MART-1 antigen (Melanoma Antigen Recognized by T cells-1). Of the ten HLA-A2 restricted TIL lines generated nine recognized MART-1, and four recognized a form of gp100 isolated and described herein (see Example 3) and none appeared to recognize MAGE-1 (Zakut, R., et al. (1993), Cancer Res 53:5-8.; data not shown).
  • TABLE 2
    Interferon Gamma Secretion by TIL501.A42 and
    TIL 1235 When Cultured with HLA-A2+ Cell Lines
    Transfected with the Gene 22 or 23
    TIL501.A42 TIL1235
    Stimulator Cell Transfected Gene HLA-A2 pg/ml
    Exp. 1
    501mel none + 1009* 1076
    397mel none <50 <50
    COS7 none <50 <50
    COS7 HLA-A2.1 + <50 <50
    COS7 22 <50 <50
    COS7 HLA-A2.1+22 + 771 1049
    Exp. 2
    501mel none + ND+ 1051
    397mel none ND <50
    293 HLA-A2.1 + ND <50
    293 22 ND <50
    293 HLA-A2.1+22 + ND 255
    Exp. 3
    501mel none + 1073  1056
    397mel none <50 <50
    MDA231 none + <50 <50
    MDA231 23 + 674 725
    A375 none + <50 <50
    A375 23 + 264 131
    IFN-γ in the supernatant was measured after TIL were coincubated for 20 hr with COS7 or 293 cell lines transiently transfected with the pcDNA3 containing the HLA-A2.1 and/or cDNA 22 by the DEAE-dextran method (Exp. 1 & 2), or with the A375 or MDA231 cell lines stably transfected with cDNA 23 (Exp. 3).
    IFN-γ was secreted only when TIL were incubated with HLA-A2+ cell lines transfected with the cDNA 22 or 23.
    *IFN-γ secreted by TIL alone without stimulator (<50 pg/ml) was subtracted.
    +Not done
  • TABLE 3
    Expression of the MART-1 Gene in a Variety of Tissue and Cell Lines
    Melanoma
    HLA-A2+
    501mel +
    526mel +
    624mel +
    Malme3M +
    952mel +
    697mel +
    C32 +
    RPMI7951
    WM115
    A375
    HLA-A2−
    397mel +
    888mel +
    537mel +
    586mel +
    Melanocyte
    NHEM483 +
    NHEM493 +
    NHEM529 +
    NHEM530 +
    FM902 +
    FM906 +
    HA002 +
    Normal fresh tissue
    Brain
    Retina +
    Adrenal gland
    Lung
    Liver
    Kidney
    Spleen
    Thymus
    Testis
    Fetal liver
    Non-melanoma cell lines
    T-cell
    TILA
    TILB
    B-cell
    Daudi
    HMY-CIR
    501EBVB
    836EBVB
    Fibroblast
    M1 fibroblast
    Kidney epithelial cell
    KAM
    WLC
    Colon cancer
    Collo
    SW480
    WiDr
    Breast cancer
    MDA231
    MCF7
    HS578
    ZR75
    Neuroblastoma
    SK-N-AS
    SK-N-SH
    Ewing sarcoma
    TC75
    RD-ES
    6647
    Sarcoma
    143B
    Glioma
    U138MG
    HS683
    Renal cell cancer
    UOK108
    UOK117
    Small cell lung cancer
    H1092
    Northern blot analysis with 10-20 ug of total RNA was probed with the full length cDNA of the gene 22. The RNA from most melanomas, all melanocyte cell lines tested and retina were positive.
  • Expression of MART-1
  • Northern blot analysis of a variety of cell lines including melanoma, melanocyte and nonmelanoma cancer cell lines and normal tissues was performed to evaluate the expression of the gene coding for MART-1 (Table 3). Seven of ten HLA-A2+ melanoma cell lines, all four HLA-A2 melanoma cell lines, and all seven melanocyte cell lines tested were positive for MART-1 RNA expression. In this Northern analysis, all HLA-A2+ melanoma cell lines recently established in our laboratory expressed MART-1 RNA. There was a perfect correlation between MART-1 expression and lysis by TIL501.A42 in the 10 HLA-A2+ melanoma lines shown in Table 3. TIL 501.A42 which recognized the MART-1 Ag lysed 13 of 17 (76%) HLA-A2+ melanoma cell lines tested (data not shown). Of ten normal human tissues examined for mRNA expression by Northern blot analysis only retina was positive. No positivity was seen in any cell lines from T cells, B cells, kidney epithelial cells or fibroblasts or in 19 nonmelanoma tumors. It thus appears that MART-1 is a previously undescribed antigen expressed on melanocyte lineage cells from skin and retina that is also expressed on melanoma cells.
  • Studies using a panel of T cell clones and immunoselected melanoma clones (Knuth, A., et al. (1989), Proc. Natl. Acad. Sci. (USA) 86:2804-2808; Wolfel, T., et al. (1987), J. Exp. Med. 170:797-810), as well as studies analyzing HPLC fractionated peptides from melanoma cells (Slingluff, C. L., et al. (1993), J. Immunol. 150:2955-2963; Storkus, W. J., et al. (1993), J. Immunol. 151:3719-3727) suggest that multiple antigenic peptides that can provoke an immune response exist on melanomas. By cDNA cloning, two genes encoding melanoma antigens have been identified; MART-1 (FIG. 1; SEQ ID NO. 1) and a gp100 gene (see Example 3; FIG. 4; SEQ ID NO. 26). MART-1 and the form of gp100 identified herein (FIGS. 4 & 5A; SEQ. ID NOS. 26 and 27), both are recognized by HLA-A2.1 restricted TIL. The MART-1 antigen is a 118 amino acid protein of approximately 13 kd. Neither the gene nor the amino acid sequence for MART-1 have been previously described.
  • MART-1 RNA was expressed in 11 of 14 HLA-A2.1 positive or negative melanoma lines, and 7 of 7 melanocyte lines. With the exception of retinal tissue no MART-1 expression was found on any normal tissue tested, T-cell lines, B-cell lines, kidney epithelial lines, a fibroblast line or 19 tumor cell lines from cancers of the colon, breast, brain, kidney, lung or bone.
  • Another melanoma antigen, MAGE-1, has been described that is recognized by T cells derived from peripheral blood lymphocytes following repetitive in vivo or in vitro immunization (Van Der Bruggen, et al. (1991), Science 254:1643-1647).
  • The identification of genes associated with melanoma tumor antigens opens new possibilities for active specific immunization approaches to the immunotherapy of patients with cancer based on the introduction of these genes into viral or bacterial vector systems. The possibility exists that immune reactions induced against melanocyte-melanoma lineage antigens such as MART-1 may be generated against normal cells. Vitiligo, probably resulting from anti-melanocyte immune reactions, has been reported to be associated with a favorable prognosis in patients with melanoma (Nordlund, J. J., et al. (1983), J. Am. Acad. Dermatol. 9:689-695); Bystryn, J-C, et al. (1987), Arch. Dermatol. 123:1053-1055), and has also been reported in patients responding to chemoimmunotherapy (Richards, J. M., et al. (1992), J. Clin. Oncol. 10:1338-1343). TIL with anti melanocyte-melanoma reactivities have been administered to patients with advanced melanoma (Rousenberg, S. A., et al. (1988), N Engl J Med 319:1076-1680; Rosenberg S. A., J. Clin. Oncol. 10:180-199) and although sporadic vitiligo has been seen in these patients, no adverse ophthalmologic effects related to the possible expression of these melanocyte antigens on retinal cells has been observed.
  • Because HLA-A2 is present in about 50% of individuals and the HLA-A2 restricted MART-1 antigen also appears to be widely expressed on melanomas, immunization with the MART-1 antigen may be particularly useful for the development of active immunotherapies.
  • EXAMPLE 2 Characterization of Immunogenic Epitopes of MART-1
  • Generation of Melanoma Specific CTL Lines and a Clone from TIL
  • Melanoma specific CTL lines were generated by culturing a single cell suspension made from metastatic melanoma with 6000 U/ml of IL2 (Cetus-Oncology Division, Chiron Corp. Emeryville, Calif.) as previously reported (Kawakami, Y. et al., (1988) J. Exp. Med. 168:2183) A T-cell clone, A42 was established by limiting dilution methods from patient, 501.
  • Assessment of antigen recognition by CTL 51Cr release cytotoxicity assays and cytokine release assays using ELISA to measure IFN-γ, GM-CSF and TNF-α were performed to analyze the reactivity of TIL as described in Kawakami, Y. et al. (1988), J. Exp. Med. 168:218 (see Example 1). Melanoma cell lines were established in the laboratory. For analysis of the recognition of known antigens by TIL, the COS7 cell line transfected with cDNAs encoding either, MART-1, gp100, or tyrosinase related protein (gp75) (Cohen, T. et al., (1990) Nucleic Acids Research 18:2807) along with HLA-A2.1 cDNA were incubated with TIL for 20 h and the amount of IFN-γ secreted into the supernatant was measured by ELISA as described in Example 1. The cDNA encoding MART-1 (see Example 1) or gp100 (see Example 3) in plasmid pcDNA3 (Invitrogen, San Diego, Calif.) was cloned from a 501mel melanoma cDNA library by screening with TIL1235 or TIL1200, respectively (see Example 1). The cDNA encoding tyrosinase related protein(gp75) in pCEV27 plasmid was isolated from 501mel melanoma cDNA library using a probe generated by PCR based on the reported sequence of gp75 (Cohen et al. (1980) Nucleic Acids Research 18:2807).
  • Peptide Synthesis and Identification of Antigenic Peptides
  • Peptides were synthesized by a solid phase method using a Gilson AMS 422 multiple peptide synthesizer. The peptides were purified by HPLC on a Vydac C-4 column with 0.05% TFA/water-acetonitrile. To identify the antigenic peptides, TIL lysis of T2 cell lines preincubated for 2h with each peptide was measured using a 51Cr release cytotoxicity assay.
  • HA-A2 Restricted Melanoma Specific TIL
  • HLA-A2 restricted melanoma specific CTL lines and a clone, A42, were established from lymphocytes infiltrating into tumors of 10 melanoma patients. These TIL recognized autologous and most. allogeneic fresh or cultured melanoma cells expressing HLA-A2, but did not recognize HLA-A2 melanomas or HLA-A2+ non-melanoma cell lines (Kawakami et al. (1992) J. Immunol 148:638). They also recognized HLA-A+ normal cultured melanocytes derived from neonatal skin (see Example 1 and Kawakami, Y. et al. (1993), J. Immunotherapy 14:88). Thus, these TIL recognized non-mutated self-peptides derived from proteins expressed in melanoma and melanocytes in association with HLA-A2.
  • Recognition of Additional Melanoma Proteins by TIL
  • To evaluate the frequency of recognition of 4 isolated melanoma proteins including MART-1, a form of gp100 (FIG. 5A; SEQ ID NO: 26, see Example 3), and tyrosinase related protein(gp75), the reactivity of TIL to COS7 was tested on cell lines transfected with cDNAs encoding these 3 proteins with or without the cDNA encoding HLA-A2.1. One of several experiments with 9 TIL is shown in Table 4. Eight of the nine HLA-A2 restricted melanoma specific TIL secreted IFN-γ when incubated with COS7 cotransfected with MART-1 and HLA-A2.1. Only TIL1200 which is a relatively oligoclonal CTL line (Shilyansky, J. et al., (1994) Proc. Natl. Acad. Sci. (USA) 91:2829) did not respond to this COS transfectant. Four TIL (620, 660, 1143, 1200) recognized gp100 when transfected along with HLA-A2.1. TIL1200 secreted large amounts of IFN-γ compared to TIL620, 660, and 1143, suggesting that only a small subset of T-cells in these latter 3 TIL lines recognized gp100. None of these TIL recognized gp75 using this assay. Thus, MART-1 is a common melanoma antigen recognized by most HLA-A2 restricted TIL derived from melanoma patients.
  • Identification of MART-1 Epitopes for TIL
  • To identify the MART-1 epitopes for these TIL, 23 peptides were selected based on the known peptide binding motifs to HLA-A2.1 (Falk, K. et al., (1993) Nature, 351:290; Hunt, D. F. et al. (1992), Science, 255:1261; Ruppert, J. et al., (1993) Cell 74:929), synthesized (>90% purity) and screened by testing lysis of the HLA-A2.1+ T2 cell line by TIL after incubation of the T2 line with each peptide (Table 5). The T2 cells (Cerundolo, V. et al.;
  • TABLE 4
    Recognition of melanoma antigens by HLA-A2 restricted melanoma specific TIL
    TIL TIL TIL TIL TIL TIL TIL TIL TIL
    Stimulator Transfected
    501 620 660 1074 1088 1128 1143 1200 1235
    Cell line cDNA HLA-A2 IFN-γ secretion (pg/ml)
    501mel none + 93 720 530 670 491 272 354 736 750
    397mel none 0 0 0 0 0 0 0 0 0
    COS7 none 0 0 0 0 0 0 14 0 0
    COS7 HLA-A2.1 + 0 0 0 0 0 0 0 0 0
    COS7 MART-1 0 0 0 0 0 0 12 0 0
    COS7 gp100 0 0 0 0 0 0 14 0 0
    COS7 gp75 0 0 0 0 0 0 0 0 0
    COS7 HLA-A2.1 + MART-1 + 270 196 131 625 328 52 184 0 743
    COS7 HLA-A2.1 + gp100 + 0 89 17 0 0 0 41 391 8
    COS7 HLA-A2.1 + gp75 + 0 0 0 11 0 0 7 0 0
    IFN-γ was measured in the supernatants after HLA-A2 restricted melanoma specific TIL were coincubated with COS7 cells cotransfected with cDNAs encoding proteins expressed in melanoma with or without HLA-A2.1 cDNA. All TIL except TIL1200 secreted IFN-γ when cultured with COS7 cotransfected with cDNAs encoding MART-1 and HLA-A2.1. TIL620, 660, 1143 and 1200 secreted IFN-γ when cultured with COS7 cotransfected with the cDNAs encoding gp100 and HLA-A2.1.

    (1990) Nature 345: 449-452) cell line was lysed well by all 4 HLA-A2 restricted melanoma specific TIL tested when preincubated with either peptides M9-2, M10-3, or M10-4. Both 10 amino acid peptides, M10-3 and M10-4 contain the M9-2 sequence, with M10-3 having an additional glutamic acid at its N-terminus and M10-4 having an extra isoleucine at its C-terminal end. These peptides are located in a hydrophobic putative transmembrane domain in MART-1. The same lysis was observed when other HLA-A2+ cells incubated with these peptides were used as targets including the K4B (provided by Dr. William Biddson, NIH; Storkus. W et al. (1993) J. of Immunology 151:3719-3727) and 501EBVB Epstein-Barr virus transformed B cells (Topalian et al. (1989) J. Immunol. 142: 3714-3725) or HMY-C1R B cells (Dr. William Biddson; NIH; Storkus, W. et al., (1993) J. of Immunol. 151:3719-3727) transfected with the HLA-A2.1 gene (data not shown).
  • The peptides, M9-1, M9-2, M9-3, M10-2, M10-3, M10-4 and M10-5 were further purified and titrated in order to evaluate their relative ability to sensitize T2 cells to lysis by MART-1 reactive TIL1235 or T cell clone A42 (FIG. 2). The purified peptides M9-2, M10-3 and M10-4 were required in minimum concentrations of 1 ng/ml, 10 ng/ml an 1000 ng/ml, respectively. The purified M10-4 was not recognized by TIL clone A42 even at 10 ug/ml as shown in FIG. 2. M9-1, M9-3, M10-2, and M10-5 peptides were not recognized by either A42 or TIL1235.
  • TABLE 5
    Lysis of T2 cells preincubated with synthetic
    MART-1 peptides
    TIL TIL TIL
    A42 1235 660 1074
    Target Peptide % specific lysis
    501mel none 47 30 31 41
    397mel none 1 0 1 2
    T2 none −2 −3 −1 1
    T2 M9-1 TTAEEAAGI −10 −5 −5 −4
    T2 M9-2 AAGIGILTV 64 80 40 56
    T2 M9-3 GIGILTVIL 18 20 0 10
    T2 M9-4 GILTVILGV 1 −1 −3 2
    T2 M9-5 ILTVILGVL −2 −1 −5 −1
    T2 M9-6 LTVILGVLL 1 0 1 0
    T2 M9-7 TVILGVLLL −2 −3 −2 1
    T2 M9-8 VILGVLLLI 1 5 −2 −2
    T2 M9-9 ALMDKSLHV −1 −4 −8 0
    T2 M9-10 SLHVGTQCA −1 1 −8 4
    T2 M9-11 PVVPNAPPA −2 0 4 −1
    T2 M9-12 NAPPAYEKL 1 −2 0 6
    T2 M10-1 YTTAEEAAGI −4 −2 −3 3
    T2 M10-2 TAEEAAGIGI 7 11 12 15
    T2 M10-3 EAAGIGILTV 55 66 31 51
    T2 M10-4 AAGIGILTVI 34 68 29 21
    T2 M10-5 GILTVILGVL −1 2 7 10
    T2 M10-6 ILTVILGVLL 1 6 6 7
    T2 M10-7 LTVILGVLLL −2 −1 −1 2
    T2 M10-8 TVILGVLLLI −6 −1 −1 11
    T2 M10-9 RALMDKSLHV 3 5 8 11
    T2 M10-10 SLHVGTQCAL −2 −8 2 9
    T2 M10-11 SLQEKNCEPV 3 2 2 9
    Twenty-three peptides (12 9-mers and 11 10-mers) (>90% purity) were synthesized and the lysability by TIL clone A42, TIL lines TIL1235, TIL660, and TIL1074 derived from different patients was tested against HLA-A2+ T2 cells preincubated with each peptide (10 ug/ml) in a 4h-51 Cr release cytotoxicity assay at E:T ratio of 20:1 for A42 and 40:1 for other TIL lines. T2 cells were lysed well when incubated with M9-2, M10-3 and M10-4. M10-3 and M10-4 contain the entire M9-2 sequence (underlined).
  • Recognition of MART-1 Peptides by HLA-A2 Restricted TIL Established from Different Patients
  • To evaluate whether a variety of HLA-A2 restricted MART-1 specific TIL recognized the same or different epitopes in the MART-1 antigen, lysis of T2 cells (Cerundolo V., et al. (1990) Nature 345: 449-452) preincubated with each peptide was tested with TIL derived from 10 melanoma patients. A representative experiment with 10 TIL is shown in Table 6. M9-2 and M10-3 were recognized by 9 of 10 TIL (only TIL1200 were negative).as well as the A42 clone with the same pattern of lysis as COS7 cells cotransfected with cDNAs encoding MART-1 and HLA-A2.1. Only TIL620 and TIL1088 demonstrated low level of non-specific lysis of T2 cells without peptides or after the addition of irrelevant peptides, but showed significant increase of lysis of T2 cells preincubated with M9-2, M10-3, and M10-5 peptides. The recognition of M10-4 differed among the TIL, but was similar to the different reactivity to M10-4 by the T-cell clone A42 or the T-cell line TIL1235 (FIG. 2). Higher concentrations (1 ug/ml) of M10-4 were required for lysis than were required for M9-2 or M10-3. These 10 TIL and clone A42 also secreted cytokines including IFN-γ, GM-CSF and TNF-α when incubated with T2 cells preincubated with M9-2 or M10-3 (data not shown). Therefore, M9-2 or M10-3 are common epitopes recognized by a majority of HLA-A2 restricted melanoma specific TIL.
  • TABLE 6
    Recognition of MART-1 peptides by HLA-A2 restricted melanoma specific TIL
    TIL TIL TIL TIL TIL TIL TIL TIL TIL TIL clone
    Peptide 501 620 660 1074 1088 1128 1143 1200 1235 1363 A42
    Target (ug/ml) % specific lysis
    501mel none 42 49 35 32 31 19 24 41 32 43 41
    397mel none 3 16 6 1 1 4 3 3 3 4 1
    T2 none 0 7 −3 −6 7 −6 −7 −6 −7 −7 −6
    T2 M9-1 (1) 4 15 −4 1 31 1 −5 −1 1 4 3
    T2 M9-2 (1) 86 75 73 79 98 30 36 2 92 82 91
    T2 M9-2 (0.001) 52 49 23 32 81 9 6 1 10 41 63
    T2 M9-3 (1) 5 25 0 1 19 0 1 −2 0 −2 −4
    T2 M10-2 (1) 10 22 5 8 21 8 3 7 7 7 6
    T2 M10-3 (1) 84 68 68 73 79 24 27 1 42 67 62
    T2 M10-3 (0.001) 91 50 33 25 86 13 14 0 14 39 1
    T2 M10-4 (1) 83 47 16 35 80 6 3 1 14 53 0
    T2 M10-4 (0.001) 0 11 3 0 14 4 −1 −1 2 −3 −3
    T2 M10-5 (1) 4 14 1 4 13 2 3 0 3 0 2
    Lysability by TIL clone A42 and TIL lines derived from 10 patients of T2 cells preincubated with the purified peptides M9-1, M9-2, M9-3, M10-2, M10-3, M10-4, and M10-5 was tested in a 4 h-51Cr release assay at an E:T ratio of 20:1 for A42 and 40:1 for other TIL lines. Nine of ten TIL lysed T2 cells incubated with peptides M9-2 or M10-3. Seven of ten TIL lysed T2 incubated with peptide M10-4 at a concentration of 1 ug/ml.
  • The relative frequency of recognition of known melanoma proteins by T-cells derived from the TIL of ten melanoma patients has been examined. The common epitopes, M9-2 and M10-3 in the MART-1 antigen that were dominantly recognized by nine of these TIL have also been identified. The cDNA encoding MART-1 was isolated by cDNA expression cloning using TIL1235 in screening assays (See Example 1). MART-1 is a 118 amino acid protein containing a single transmembrane domain and is expressed in most melanoma cells as well as cultured melanocytes and retina similar: to the expression pattern of the cDNA for a form of gp100 described in Example 3. The gp100 is recognized by 4 of 10 TIL.
  • Based on dose response analysis, peptide M9-2 most effectively sensitized T2 cells for lysis (FIG. 2) suggesting that this peptide may be naturally processed and presented on tumor cells. The T-cells recognizing M9-2 may react with peptide M10-3 or M10-4 because the latter 10-mer peptides contain the 9 amino acid sequence of peptide M9-2. There is some difference in recognition of these 3 peptides by different TIL. For example, M10-4 was poorly recognized by the T-cell clone A42, but was well recognized by some TIL lines, although a higher concentration of M10-4 was necessary to observe the lysis. This may be due to the variation of TCR affinity for the M9-2 and M10-4 peptides in the context of HLA-A2, or alternatively, TIL lines may contain different T-cell clones which only recognize either M9-2 or M10-4. Peptides M10-3 and M10-4 may also be naturally processed and presented by tumor cells. The existence of multiple melanoma antigens presented by HLA-A2 has previously been suggested by analyzing the recognition of melanoma cell clones by a variety of T-cell clones (Knuth, A. et al. (1989), Proc. Natl. Acad. Sci. (USA) 86:2804, Wolfel, T. et al., 1989 J. Exp. Med. 170:797) or by analyzing HPLC peptide fractions that were isolated from HLA-A2 melanoma cells (Slingluff, C. L. Jr. et al., (1993) J. Immunol. 150:2955, Storkus; W. J. et al., (1993) J. Immunol. 151:3719).
  • The observation that most HLA-A2 restricted TIL from melanoma patients recognize common MART-1 peptides but not gp75 suggests that the M9-2 or M10-3 MART-1 peptides may be more immunogenic in inducing T-cell responses in vivo than other known melanoma antigens. Some of the TIL used in this study were injected along with IL2 into autologous patients, and interestingly, all 4 TIL (620, 660, 1074, 1200) that recognize a gp100 protein (FIG. 5A; SEQ ID NO: 27) effectively induced tumor regression (more than 50% reduction of tumor). All but TIL1200 also recognized MART-1.
  • EXAMPLE 3 Identification of a Second Human Melanoma Antigen Recognized by Tumor Infiltrating Lymphocytes Associated with in Vivo Tumor Rejection cDNA Expression Cloning
  • The cDNA25 clone encoding a form of the melanoma antigen designated gp100 was cloned by techniques similar to those described in Example 1 and in Miki, T., et al. (1991) Proc. Natl. Acad. Sci. (USA) 88:5167-5171. Briefly, a breast cancer cell line, MDA231 (ATCC #HTB26), transfected with a cDNA library in λpCEV27 made from the 501mel melanoma cell line was screened for antigen positivity by measuring interferon-λ (IFN-γ) secretion when cocultured with TIL1200. TIL1200 was generated as described in Kawakami, Y., (1988), J. Exp. Med. 168, 2183-2191. The integrated cDNA was recovered from the genomic DNA of positive transfectants by PCR and cloned into the mammalian expression plasmid pcDNA3 (Invitrogen, San Diego, Calif.). The full length cDNA for cDNA25 was isolated from the 501mel λpCEV27 library using the cDNA25 probe. The λphage containing the full length cDNA25 was digested with XhoI, and then self-ligated with T4 DNA ligase to make the plasmid pCEV27-FL25. Alternatively, a full length cDNA25 isolated by PCR using the specific primers designed for gp100 was cloned in pCRII(Invitrogen), and then cloned into pcDNA3(pcDNA3-FL25). To test whether this cDNA encoded a melanoma antigen it was retransfected into COS7, A375 or MDA231 and the resulting transfectants were tested for stimulation of TIL1200. DNA sequences of the plasmid clones were determined with an automated DNA sequencer (Model 373A; Applied Biosystems, Inc.), using the Taq DyeDeoxy terminator cycle sequencing kit (Applied Biosystems, Inc.) using the manufacturer's instructions.
  • Peptide Synthesis and Identification of Antigenic Peptides
  • Peptides were synthesized by a solid phase method using a Gilson AMS 422 multiple peptide synthesizer. The peptides were purified by HPLC on a Vydac C-4 column with 0.05% TFA/water-acetonitrile. To identify antigenic peptides, TIL lysis of T2 RET-cells preincubated with peptides for 2 hour (h) was measured using a 51Cr release cytotoxicity assay.
  • Treatment of a Patient with Metastatic Melanoma Using TIL 1200
  • A 29 year old male patient, designated patient number 1200, with a widely metastatic melanoma who had previously failed chemotherapy and radiation therapy was treated with a single preparatory dose of 25 mg/Kg cyclophosphamide followed by the intravenous infusion of 1.6×1011 TIL (including 9.1×109 Indium-ill labeled TIL) plus 7 doses of IL-2 at 720,000 IU/Kg given every 8 hours. A second cycle of treatment with TIL and IL-2 was given three weeks later. Radionuclide scans showed localization of TIL in rumor deposits (FIG. 3A). Biopsy of subcutaneous tumors on days 8 and 11 after treatment showed significant localization of TIL to tumor (ratios of injectate per gram in tumor compared to normal tissue were 14.9 and 14.0 respectively). The patient's cancer regressed rapidly following the first course of treatment. By three months after treatment 2 of 3 liver lesions had disappeared and a third lesion shrank by 50%. Multiple subcutaneous metastases regressed completely as shown in FIG. 3B (the product of perpendicular diameters of individual lesions are shown).
  • Characterization of in vitro function of TIL1200 A number of TIL lines established from HLA-A2+ melanoma patients lysed melanoma cell lines in a class I MHC-restricted fashion (Kawakami, Y., et al. (1992) J. Immunol. 148; 638-643), and were shown to release IFNγ, tumor necrosis factor-alpha (TNFα) or granulocyte-macrophage colony stimulating factor (GM-CSF) when cocultured with the same tumor cell lines (Hom, S-S., et al. (1993) J. Immunother. 13; 18-30). A CD8+ CTL line, TIL1200, established from a metastatic subcutaneous tumor mass of patient 1200, lysed fresh autologous melanoma cells as well as 10 of 15 HLA-A2+ allogeneic melanoma cell lines, but did not lyse 16 of 18 HLA-A2 melanoma cell lines or 6 of 8 HLA-A2+ non-melanoma cell lines (Shilyansky, J., et al. (1993) Proc. Natl. Acad. Sci. USA, 91, 2829-2833, unpublished data). Table 7 shows a cytotoxicity assay against 5 representative HLA-A2+ melanoma cell lines that were lysed by TIL1200, 4 representative HLA-A2+ melanoma cell lines that were not lysed by TIL1200, and one HLA-A2 melanoma cell line. TIL1200 also secreted IFN-γ when cocultured with HLA-A2+ normal cultured melanocytes established from neonatal foreskin as well as HLA-A2+ melanoma cell lines (Table 8). Therefore, TIL1200 appeared to recognize a non-mutated self peptide expressed in most melanomas and cultured neonatal melanocytes in an HLA-A2 restricted fashion.
  • Cloning of the cDNA Coding for a Melanoma Antigen Recognized by T Cells
  • A cDNA library in λ pCEV27 from the HLA-A2+ 501mel melanoma cell line, which was lysed by most HLA-A2 restricted melanoma specific TIL, was stably transfected into the highly transfectable HLA-A2+ melanoma antigen negative MDA231 clone 7 or A375 clone 1-4. G418 resistant cells were selected and approximately 6700 individual transfectants from each cell line were isolated and screened based on their ability to stimulate IFN-γ secretion from TIL1200. Six DNA fragments were isolated by PCR using SP6/T7 primers flanking the integrated DNA from four MDA231 and one A375 transfectants that were positive in a second screening and were cloned into the mammalian expression vector pcDNA3 (Invitrogen).
  • These fragments in the pcDNA3 vector were transiently expressed in the COS7 cells with or without pcDNA3-HLA-A2.1. Transfection into COS7 of one of the cDNAs tested, cDNA25, along with HLA-A2.1 reproducibly conferred the ability to stimulate secretion of IFN-γ from TIL1200. The stable transfection of cDNA25 into A375 also stimulated IFN-γ release from TIL1200 (Table 9, Exp.1 and Exp. 2). A 2.2 Kb band detected by Northern blot analysis of the melanoma using the cDNA25 probe suggested that the cloned 1.6 Kb fragment was not a full length cDNA. Comparison with the GenBank database of the consensus DNA sequence of
  • TABLE 7
    Specificity of Antigen Recognition by TIL1200:
    Lysis of HLA-A2+gp100+ Melanoma Cell Lines
    TIL1200 LAK
    gp100 (% specific
    Target HLA-A2 FACS Northern lysis)
    501mel + + + 46 78
    526mel + + + 39 74
    624mel + + + 33 76
    952mel + + + 25 76
    Malme3M + + + 43 70
    C32 + −/+* 6 82
    RPMI7951 + 9 67
    WM115 + 5 68
    HS695T + 3 87
    397mel + + 0 70
    5 hour (h) 51Cr release assay was performed to measure cellular cytotoxicity at an effector: target ratio of 40:1 as previously described(Kawakami, Y. et al. (1988) J. Exp. Med. 168: 2183-2191). The expression of HLA-A2 and gp100 recognized by monoclonal antibody HMB45 (Enzo Diagnostics, New York, NY) was measured by flow cytometry(FACS). The expression of gp100 RNA was analyzed by Northern blot with a cDNA25 probe.
    *−/+ indicates very weak positive.
  • TABLE 8
    Specificity of Antigen Recognition by TIL1200:
    Recognition of HLA-a-a2+ Neonatal Melanocytes
    TIL1200 TIL888
    Stimulator HLA-A2 (pgIFN-γ/ml)
    501mel + 562 0
    624mel + 439 0
    397mel 0 0
    888mel 0 1970
    NHEM493 441 0
    NHEM527 + 418 0
    NHEM530 + 164 0
    NHEM616 + 53 0
    FM725 + 107 0
    FM801 + 250 343
    NHEM483 0 0
    NHEM680 0 0
    HA002 0 0
    The IFN-γ secretion by TIL was measured by ELISA as previously described in Example 1. The amount of IFN-γ secreted by TIL alone was subtracted (88 pg/ml for TIL888 and none for TIL 1200). TIL888 is a class I MHC restricted melanoma specific CTL, not restricted by HLA-A2. NHEM, FM, and HA refer to normal cultured melanocyte cell lines, all others are melanoma cell lines.
  • TABLE 9
    Transfection of cDNA 25 into A375 and COS7
    Secretion by
    Stimulator Transfected HLA- TIL1200
    cells genes A2 (pgIFNγ/ml)
    Exp. 1 501mel none + 987
    397mel none 0
    A375 none + 0
    A375 pcDNA3-25 + 230
    Exp. 2 501mel none + 662
    397mel none 0
    COS7 none 0
    COS7 HLA-A2.1 + 0
    COS7 pcDNA3-25 0
    COS7 HLA-A2.1+pcDNA3-25 + 310
    Exp. 3 501mel none + 908
    397mel none 0
    COS7 none 0
    COS7 HLA-A2.1 + 0
    COS7 pCEV27-FL25 0
    COS7 HLA-A2.1+pCEV27-FL25 + 742
    COS7 pcDNA3-FL25 0
    COS7 HLA-A2.1+pcDNA3-FL25 + 801
    TIL 1200 secreted IFN-γ when coincubated with HLA-A2+ A375 stably transfected with pcDNA3 containing truncated cDNA25 (pcDNA3-25) (Exp. 1) or COS7 transiently transfected with either pcDNA3-25 (Exp. 2), pcDNA3 containing full length cDNA25 (pcDNA3-FL25) or pCEV27 containing full length cDNA25 (pCEV27-FL25) (Exp. 3) along with pcDNA3 containing HLA-A2.1 (HLA-A2.1). HLA-A2 expression was determined by flow cytometry and interferon-gamma secretion was measured by ELISA.

    3 cDN25 clones that were independently amplified by PCR revealed that cDNA25 was distinct from two previously registered genes, a gp100 (GenBank Access No. M77348) and Pmel17 (Kwon, B. S., et al. (1991) Proc. Natl. Acad. Sci, USA 88, 9228-9232). The cDNA 25 differed from the gp100 in GenBank (Accession No. M77348, also known as gp95) by two nucleotides, from the PMEL 17 sequence (Kwon et al. (1991) Proc. Natl. Acad. Sciences (USA) 58: 9228-9232) by 3 bases and a 21 base pair deletion. (FIG. 5B).
  • The full length cDNA25(FL25) was isolated in two plasmids, pCEV27-FL25 or pcDNA3-FL25. Transfection of either plasmid into COS7 along with pcDNA3-HLA-A2.1 conferred to COS7 the ability to induce IFN-γ secretion by TIL1200. The amount of IFN-γ secretion stimulated by COS7 transfected with the full length DNA plus HLA-A2.1 was similar to that stimulated by 501mel and was higher than that stimulated by COS7 transfected with the truncated cDNA25 possibly due to improved translation starting at the normal AUG initiation codon (Table 9, Exp.2 and 3). Alternatively, the 5′ region missing from the truncated cDNA25 may contain other epitopes recognized by clones in TIL1200. The requirement for HLA-A2.1 expression for IFN-γ release from TIL1200 and the fact that transfected cells did not stimulate IFN-γ secretion from irrelevant TIL (data not shown) demonstrated that the cDNA25 encoded an antigen recognized by TIL1200 in the context of HLA-A2.1 and did not encode a molecule that non-specifically induced IFN-γ release from T cells.
  • The nucleotide and corresponding amino acid sequences of the truncated cDNA 25 and the full length cDNA25 cloned from the 501mel cDNA library by screening with the cDNA25 probe (FIG. 5A) were compared with the GenBank sequences of Pmel17 isolated from normal melanocytes and gp100 isolated from the melanoma cell line MEL-1. (FIG. 5B). The full length cDNA25 differed from the gp100 amino acid sequence at position 162. This amino acid difference is possibly caused by polymorphism or mutation in the tumor. cDNA25 had 2 amino acid differences at positions 162 and 274, compared to Pmel17 and did not contain 7 amino acids that existed in Pmel17 at positions 588-594. The amino acid sequence of the truncated cDNA25 that was isolated from the original MDA231 transfectant has a different sequence at the 3′ end (from position 649 to the end) due to a frame shift caused by one extra cytidylic acid. It is not clear whether this difference was due to a true allelic difference or to a mutation that occurred during manipulation of the DNA. Nevertheless, TIL1200 appeared to recognize non-mutated peptides located between position 236 and 648. cDNA25 also had 87% similarity in amino acid sequence to cDNA RPE1 (Kim, R., and Wistow, G. J. (1992) Exp. Eve Res. 55: 657-662) specifically expressed in bovine retinal pigment epithelium and 60% similarity to cDNA MMP115 that encoded a melanosomal matrix protein isolated from chicken pigmented epithelial cells (Shilyansky, J., et al. (1993) Proc. Natl. Acad. Sci, USA, 91, 2829-2833).
  • A gp100 protein was known to be recognized by monoclonal antibody HMB45 (Adema et al., (1993) Am. J. Patholoay, 143: 1579-1585). COS7 cells transfected with the full length cDNA25 were evaluated by flow cytometry using this monoclonal antibody. After transient expression of either pCEV27-FL25 or pcDNA3-FL25, COS7 expressed the antigen detected by HMB45 (data not shown).
  • Expression of RNA for cDNA25
  • Northern blot analysis was performed with the cDNA25 probe to evaluate the tissue specific expression of this gene. Ten of 15 melanoma cell lines and 6 of 6 melanocyte cell lines were positive for cDNA 25. Of many normal tissues tested only retina was positive (FIG. 6). Seven cell lines from T-cell (TILA, B), B-cells (501EBVB, 836EBVB) and fibroblast (M1) and 20 non-melanoma tumor cell lines (colon cancer, Collo, SW480, WiDr; breast cancer, MDA231, MCF7, HS578, ZR75; neuroblastoma, SK—N-AS, SK—N—SH; Ewing sarcoma, TC75, RD-ES, 6647; sarcoma 143B; glioma, U138MG, HS683; renal cell cancer, UOK108, UOK117, small cell lung cancer, H1092; Burkitt's lymphoma, Daudi; myeloma HMY) were all negative for cDNA25 (data not shown). Therefore, this gene appeared to be specifically expressed in melanocyte lineage cells, consistent with the expression pattern of previously isolated forms of gp100 when analyzed using monoclonal antibodies, HMB45, NKI/betab, or HMB-50 (Adema, G. J., et al. (1993) Am J Pathology 143: 1579-1585; Gown, A. M., et al., (1986) Am J Pathol 123:195-203; Colombari, R., et al. (1988) Virchows Archiv A Pathol Anat. 413:17-24); Vennegoor, C., et al. (1988) Am. J. Pathol. 130:179-192; Vogel, A. M., and Esclamado R. M. (1988) Cancer Res. 48:1286-1294). The levels of expression of the RNA detected by the cDNA25 probe in cultured neonatal melanocyte cell lines was significantly lower than that in melanoma cell lines. There was a perfect correlation between gp100 expression detected by Northern blot analysis with cDNA25 and flow cytometry using HMB45 antibody and melanoma lysis by TIL1200 in the 10 HLA-A2+ melanoma cell lines as shown in Table 7.
  • Identification of the Epitope in gp100
  • Based on a comparison of the amino acid sequence of the truncated form of cDNA 25 to known binding motifs of HLA-A2.1 (Falk, K., et al. (1992) Nature 351:290-296; Hunt, D. F., et al. (1992) Science 255:1261-1263; Ruppert, J., et al. (1993) Cell 74:929-9937,) 30 peptides of 9 or 10 amino acids in length from cDNA25 were synthesized. TIL1200 lysed the HLA-A2+ cell line, T2, only when incubated with the peptide LLDGTATLRL (SEQ ID NO: 27 residues 457-486. FIG. 5A; SEQ ID NO: 33) but not when incubated with the other 29 peptides (Table 10, FIG. 5A). Only peptide LLDGTATLRL (SEQ ID NO: 33) was able to also stimulate IFN-γ secretion by TIL 1200 (data not shown).
  • Many melanoma-specific CTL derived from TIL appear to recognize non-mutated self peptides derived from melanocyte-melanoma lineage specific proteins, since these TIL recognize most melanoma cell lines and normal cultured melanocytes sharing the appropriate restriction element. (Anichini, A., et al. (1993) J. Exp. Med. 177:989-998; Kawakami, Y., et al. (1993) J. Immunother. 14:88-93). In an attempt to isolate and identify melanoma antigens of value in the immunotherapy of melanoma patients, TIL, TIL1200 were used that, when transferred into a patient with metastatic cancer, localized to the tumor site and was associated with a dramatic tumor regression. It has been shown that, in contrast to non-activated lymphocytes and lymphokine activated killer cells, autologous TIL localize to tumor sites. This localization correlated with the ability of these TIL to mediate tumor regression (data not shown). TIL1200 which was a TIL line containing multiple CTL species recognized a tumor antigen in the context of HLA-A2, which is the most frequently expressed class I MHC antigen (about 50% of individuals) and has been shown to be a dominant restriction element for the induction of melanoma specific CTL. (Crowley, N. J., et al. (1991) J. Immunol. 146, 1692-1699).
  • By cDNA expression cloning using T cell recognition for screening, a cDNA (FIG. 4; SEQ ID NO: 26) encoding an antigen recognized by TIL1200 and identified as a form of gp100, a membrane glycoprotein also recognized by monoclonal antibodies, BMB45, HMB50 or NKI/betab has been identified. (Adema, G. J., et al. (1993) Am J Pathology 143, 1579-1585. Gown, A. M., et al. (1986) Am J Pathol 123, 195-203. Colombari, R., et al. (1988) Virchows Archiv A Pathol Anat. 413, 17-24; Vennegoor, C., et al. (1988) Am. J. Pathol. 130, 179-192; Vogel, A. M., and Esclamado R. M. (1988) Cancer Res. 48, 1286-1294). These antibodies are highly specific for melanocyte lineage tissues and strongly stain most melanoma cells. NKI/betab also reacts with adult melanocytes in normal skin (Vennegoor, C., et al. (1988)
  • TABLE 10
    TIL1200 Lysis of the T2 HLA-A2+ Cell Line
    Pulsed with the Peptide, LLDGTATLRL
    Peptide* TIL1200 TIL1235+
    Target HLA-A2 (ug/ml) (% specific lysis)**
    501mel + 0 66 51 
    397mel 0 1 0
    T2 + 0 2 1
    T2 + 40 28 ND++
    T2 + 10 32 0
    T2 + 1 24 ND
    T2 + 0.1 6 ND
    T2 + 0.01 0 ND
    T2 + 0.001 2 ND
    *TIL1200 lysed T2 cells pulsed with the 10-mer peptide, LLDGTATLRL(457-466), but not other 29 peptides SEQ ID NO: 27 (residues 273-281, 297-306, 373-381, 399-407, 399-408, 409-418, 456-464, 463-471, 465-473, 476-485, 511-520, 519-528, 544-552, 544-553, 570-579, 576-584, 576-585, 585-593, 592-600, 597-605, 597-606, 602-610, 602-611, 603-611, 605-614, 606-614, 606-615, 619-627, 629-638)
    +TIL1235 is an HLA-A2 restricted melanoma specific CTL that does not recognize gp100.
    **E:T of 50:1
    ++ND, not done.

    Am. J. Pathol. 130, 179-192) Immunoelectron-microscopic studies using either HMB45 or NKI/betab antibody revealed that a gp100 protein was mainly located in a membrane and filamentous matrix of stage I and II melanosomes in the cytoplasm (Vennegoor, C., et al. (1988) Am. J. Pathol. 130, 179-192; Schaumburg-Lever, G., et al. (1991) J. Cutan. Pathol. 18, 432-435). By a completely independent procedure, a cDNA encoding another form of gp100 was also isolated by screening with a rabbit polyclonal antiserum against gp100 (Adema, G. J., et al. (1993) Am J Pathology 143:1579-1585) and TIL1200 also lysed HLA-A2+ cell lines transfected with this cDNA clone (Bakker, A. B. H. et al. (1994) J. Exp. Med. 179:1005-1009).
  • The existence of T cells reactive to the self-antigen gp100 in tumors and the possible enrichment of these T cells at the tumor site as a possible consequence of the specific accumulation and expansion of antigen reactive cells (Sensi, M. et al., (1993) J. Exp. Med 178:1231-1246) raises important questions about the nature of the immune response to self antigens on growing cancers and about the mechanisms of immunologic tolerance to self-antigens. The increased expression of gp100 on melanoma cells relative to that in melanocytes demonstrated by Northern blot analysis or the unique inflammatory conditions that might exist at the tumor site, which may be associated with the secretion of cytokines and expression of costimulatory molecules on the cell surface, could break tolerance to gp100. Depigmentation has been reported to be associated with a good prognosis (Nordlund, J. J., et al. (1983) J. Am. Acad. Dermatol. 9:689-695; Bystryn, J-C, et al. (1987) Arch. Dermatol., 123:1053-1055) and with clinical response to chemoimmunotherapy (Richards, J. M., et al. (1992) J. Clin. Oncol. 10:1338-1343) in melanoma patients. Sporadic vitiligo in has been seen patients receiving melanoma-specific TIL but adverse ophthalmologic effects that might be related to melanocyte destruction has not been observed. Patient 1200 did not develop vitiligo or any ophthalmologic side-effects.
  • The gp100 protein (FIG. 5A; SEQ ID NO: 27) and the ten amino acid peptide identified may represent a human tumor rejection antigen since the transfer into patient 1200 of TIL1200 plus IL2 was associated with cancer regression. The traffic of TIL1200 to tumor deposits in vivo and the rapidity of the antitumor response are characteristics of the response to TIL therapy, although IL2 may also have been involved in the tumor rejection. Adoptive transfer of 3 other TIL lines which recognized gp100 as well as MART-1 also mediated tumor regression (data not shown).
  • Tyrosinase (Brichard, V., et al. (1993) J. Exp. Med. 178, 489-495) and MART-1 (see Example 1) have been identified as melanoma antigens recognized by HLA-A2 restricted CTL. Another antigen, MAGE-1 is recognized by HLA-A1 restricted melanoma-specific CTL and is expressed on a variety of cancer cells as well as testis (Van Der Bruggen, P. et al. (1991) Science, 254:1643-1647). However, none of the ten HLA-A2 restricted TIL recently developed appeared to recognize MAGE-1 (Zakut, R., et al. (1993) Cancer Res. 53: 5-8).
  • The wide expression of gp100 proteins in melanomas, the recognition of a peptide by T cells infiltrating into tumor, its restriction by HLA-A2, present in 50% of individuals, and the association of anti gp100 reactivity with cancer regression in patient 1200 imply that the gp100 antigen in particular the novel immunogenic peptides derived from the gp100 amino acid sequence (FIG. 5A; SEQ ID NO: 27) may be particularly useful for the development of active immunotherapies for patients with melanoma.
  • EXAMPLE 4 Recognition of Multiple Epitopes in Human Melanoma Antigen by TIL Associated with in Vivo Tumor Recognition Materials and Methods Generation of CTL from TIL and Treatment of Patients with Metastatic Melanoma
  • Melanoma specific CTL were induced and expanded from TIL in media containing 6000 IU/ml of IL2 as previously described (Kawakami, et al., (1988) J. Exp. Med. 168:2183). All available HLA-A2 restricted melanoma specific CTL which were administered to autologous patients in the Surgery Branch, NCI, were used in this study. TIL were administered intravenously along with IL2 into autologous patients with metastatic melanoma as previously reported (Rosenberg, S. A., et al., (1988) N Engl J Med 319:1676; Rosenberg S. A., et al., (1994) J. NCI. 86:1159). Fisher's exact test was used to determine the association of gp100 recognition by TIL with clinical response to TIL treatment; likewise with MART-1 recognition.
  • Synthesis of Peptides
  • Peptides were synthesized by a solid phase method using a peptide synthesizer (model AMS 422;Gilson Co.Inc., Worthington, Ohio)(>90% purity). The peptides to be synthesized were selected from the reported human sequence of gp100 based on HLA-A2.1 binding motifs (Falk, K., (1991) Nature 351:290; Hunt, D. F., et al, (1992) Science 255:1261; Ruppert, J., et al., (1993) Cell 74:929; Kubo, R T, et al. (1994) J Immunol. 152:3913). The following peptides were tested: Eight 8-mer peptides (with residues starting at −199, 212, 218, 237, 266, 267, 268, 269; see FIG. 7A), eighty-four 9-mer peptides with residues starting at −2, 4, 11, 18, 154, 162, 169, 171, 178, 199, 205, 209, 216, 241, 248, 250, 255, 262, 266, 267, 268, 273, 278, 280, 273, 286, 287, 296, 290, 309, 316, 332, 335, 350, 354, 358, 361, 371, 373, 384, 389, 397, 399, 400, 402, 407, 408, 420, 423, 425, 446, 449, 450, 456, 463, 465, 485, 488, 501, 512, 536, 544, 563, 570, 571, 576, 577, 578, 583, 585, 590, 592, 595, 598, 599, 601, 602, 603, 604, 606, 607, 613, 619, 648; see FIG. 7A) and seventy-seven, 10-mer peptides with residues starting at −9, 17, 57, 87, 96, 154, 161, 169, 177, 197, 199, 200, 208, 216, 224, 232, 240, 243, 250, 266, 267, 268, 272, 285, 287, 289, 297, 318, 323, 331, 342, 350, 355, 357, 365, 380, 383, 388, 391, 395, 399, 400, 406, 407, 409, 415, 432, 449, 453, 457, 462, 476, 484, 489, 492, 511, 519, 536, 543, 544, 548, 568, 570, 571, 576, 577, 584, 590, 595, 598, 599, 601, 602, 603, 605, 611, 629; see FIG. 7A) were synthesized. Possible epitopes identified in the first screening were further purified by HPLC on a C-4 column (VYDAC, Hesperia, Calif.)(>98% purity) and the molecular weights of the peptides were verified by mass spectrometry measurement as previously described (Example 3; Kawakami, Y., et al., (1994) J. Exp. Med. 180:347; Kawakami, Y., et al., (1994) Proc Natl Acad Sci (USA) 91:6458).
  • Peptide Binding Assay to HLA-A2.1
  • Soluble HLA-A2.1 heavy chain, human beta 2-microglobulin, radiolabeled peptide HBc18-27 (FLPSDYFPSV) and various concentrations of the sample peptides were coincubated in the presence of protease inhibitors for 2 days at room temperature as previously described (Ruppert, J., et al., (1993) Cell 74:929; Kubo, R T, et al., (1994). J Immunol. 152:3913; Sett A., et al., (1994). Molecular Immunol. 31:813). The percentage of labeled peptide bound to HLA-A2.1 was calculated after separation by gel filtration and the concentration of the sample peptide necessary to inhibit 50% of the binding of the labeled peptide was calculated. The relative affinity of peptides to HLA-A2.1 were also calculated as a ratio (concentration of the standard HBc18-27 peptide to inhibit 50% of the binding of the labeled peptide/concentration of the sample peptide to inhibit 50% of the binding of the labeled peptide) as previously described (Sett A., et al., (1994) Molecular Immunol. 31:813). Peptide binding was defined as high (50% inhibition at <50 nM, ratio>0.1), intermediate (50-500 nM, ratio 0.1-0.01) or weak (>500 nM, ratio<0.01) (Ruppert, J., et al., (1993) Cell 74:929; Kubo, R T, et al., (1994) J Immunol. 152:3913; Sett A., et al., (1994) Molecular Immunol. 31:813).
  • The pcDNA3 plasmid containing the full length gp100 cDNA (FIG. 4, Example 3; Kawakami, Y., et al., (1994)). Proc Natl Acad Sci (USA) 91:6458) was digested with Xho I and Xba I. After incorporation of alpha-phosphorothioate deoxynucleoside triphosphate into the Xba I site, a standard exonuclease III nested deletion was performed using the Exo Size Deletion Kit (New England Biolabs, inc., Beverly, Mass.). The deleted clones were self-ligated and amplified. The exact deletion for each clone was confirmed by DNA sequencing. To identify the region containing epitopes, pcDNA3 plasmids (Invitrogen, San Diego Calif.) containing the cDNA fragments (D3, D5, D4, C3) generated by the sequential deletion with exonuclease from the 3′ end of the full length gp100 cDNA as well as the truncated gp100 cDNA lacking the 5′-coding region (25TR) (Example 63; Kawakami, Y. (1994) Proc Natl Acad Sci (USA) 91:6458), were transfected into COS7 cells along with the HLA-A2.1 cDNA and the recognition of the transfected COS cells by TIL was evaluated using IFN-γ release assays (Example 1; Kawakami, Y., (1994) Proc Natl Acad Sci (USA) 91:3515).
  • Evaluation of Antigen Recognition by T-Cells
  • To assess antigen recognition by T-cells, a 51Cr release assay or an IFN-γ release assay were performed as previously described (Examples 1 and 2; Kawakami, Y., et al., (1994)). Proc Natl Acad Sci (USA) 91:3515; Kawakami, Y., et al., (1988). J. Exp. Med. 168:2183). Either COS7 cells transfected with cDNA encoding melanoma antigens and HLA-A2.1 cDNA, or T2 cells preincubated with peptides were used as stimulators for the IFN-γ release assay. T2 cells pulsed with peptides were also used as targets for cytotoxicity assays (Kawakami, Y., (1994) J. Exp. Med. 180:347).
  • Recognition of gp100 by TIL Correlated with Clinical Response to TIL Treatment
  • Four of 14 HLA-A2 restricted melanoma specific CTL derived from TIL recognized gp100 while 13 recognized MART-1 (3 recognized both gp100 and MART-1). None recognized tyrosinase or gp75 as assessed by the reactivity of TIL against COS7 cells transfected with the cDNA encoding these melanoma antigens along with HLA-A2.1 cDNA (Example 2; Kawakami, Y et al. (1994) J. Exp. Med. 180:347). The HLA-A2 restriction and the recognition specificity of these 4 gp100 reactive CTL has been previously demonstrated (Examples 1-3; Kawakami, Y., et al., (1994) Proc Natl Acad Sci (USA) 91:6458; Kawakami, Y., et al., (1992) J Immunol 148:638; O'Neil, B. H., et al., (1993) J Immunol 1410:1418; Shilyansky, J., et al., (1994) Proc. Natl. Acad. Sci. (USA) 91:2829). Ten of these 14 CTL were administered into the autologous patients along with IL2. As summarized in Table 11, all 4 patients treated with CTL capable of recognizing gp100 resulted in an objective partial response (>50% tumor regression). Clinical response to TIL therapy associated with reactivity of TIL to gp100 (p=0.0048) but not to MART-1 (p=0.4). These data suggested that gp100 may contain epitopes capable of mediating in vivo tumor regression.
  • Identification of Epitopes Recognized by gp100 Reactive TIL
  • To identify the epitopes recognized by these 4 gp100 reactive CTL, a 169 peptides which contained HLA-A2.1 binding motifs were synthesized. Peptide recognition was evaluated by testing the reactivity of these CTL against HLA-A2.1+ T2 cells preincubated with each peptide using both cytotoxicity and IFN-γ release assays. As shown in Table 12, 7 peptides were recognized by gp100 reactive TIL in the cytotoxicity assays. The results of the IFN-7 release assays performed at the same time were consistent with that of the cytotoxicity assays. The different subcultures of TIL620 (620-1, 620-2) or TIL660 (660-1, 660-2, 660-3) were grown from the TIL culture that was administered into the autologous patient, but they were separately cultured and had slightly different specificities likely due to the in vitro expansion of different clones. G9209 (ITDQVPFSV) (SEQ ID NO:48) and G10209 (TITDQVPFSV) (SEQ ID NO:49), which has an extra threonine at the N-terminus of G9209, were recognized only by TIL620. G9154 (KTWGQYWQV) (SEQ ID NO:46) and G10154 (KTWGQYWQVL) (SEQ ID NO:47), which has an extra leucine at the C-terminus of G9154, were recognized by TIL1200, TIL620-2 and TIL660-2. G10-4 (LLDGTATLRL) (SEQ ID NO:33) was recognized by TIL1200 as demonstrated (Example 3). The peptide G9280 (YLEPGPVTA) (SEQ ID NO:40) was recognized by TIL660 and TIL1143. TIL660-3 also recognized G10-5 (VLYRYGSFSV) (SEQ ID NO:34) as well as G9280. Lysis of T2 cells preincubated with G10-5 was repeatedly low, possibly because a small subset of T-cell clones was specific for this epitope.
  • To complement the epitope identification using the known HLA-A2.1 binding motifs, another method was also used to identify regions possibly containing epitopes. Five gp100 cDNA fragments, 4 generated by exonuclease deletion from the 3′-end of the cDNA (D3, D4, D5, C4) as well as a partial cDNA clone lacking the first 705 base pairs of the 5′-coding region (25TR), were inserted into the pcDNA3 plasmid and transfected into COS7 cells along with the HLA-A2.1 cDNA. The locations of the fragments are shown in FIG. 7A. The recognition of these transfectants by the 4 gp100 reactive TIL was evaluated using an IFN-γ release assay (FIG. 7B). TIL 1200 recognized COS cells transfected with the fragments, 25TR, D5, D4 or C4, but not with D3, suggesting that at least 2 epitopes existed in the regions of amino acid residues 146-163 and 236-661. G9154 and G10154 were the only peptides which contained HLA-A2.1 binding motifs in the region 146-163 and both were recognized by TIL1200. G10-4 was located in the region 236-661 and was recognized by TIL1200. TIL620-1 recognized COS cells transfected with C4 but not with D3, D5, D4 or 25TR, suggesting that the epitope existed within residues 187-270. G9209, and G10 208 which were recognized by TIL620-1 were located in this region. TIL620-2, another subculture of TIL620, also recognized COS cells transfected with D5 and D4, but not D3, and recognized G9154 and G20154 in the region 147-163, also recognized by TIL1200. TIL660-1 and TIL1143 recognized COS cells transfected with C4 or 25TR, but not with D3, D5, or D4, suggesting that epitopes existed in the 2 regions 187-270 and 236-661. G9280 located in the fragment 25TR, but not in the fragment C4, was recognized by TIL660 and TIL1143.
  • Binding Affinity of the Melanoma Epitopes to HLA-A2.1 in Vitro
  • With the exception of G10-4, which required a concentration of 1 ug/ml to sensitize T2 cells for CTL lysis (Example 3; Kawakami, Y., et al., (1994) Proc Natl Acad Sci (USA) 91:6458), all gp100 epitopes identified in this study could sensitize T2 cells for CTL lysis at a concentration of 1 ng/ml (FIGS. 8A-8D). G10-5 appeared to be inhibitory to the cytotoxic activity of CTL at concentration greater than 10 ng/ml since lysis of T2 cells incubated with G10-5 at more than 10 ng/ml was repeatedly lower than at 1-10 ng/ml in this assay condition in which the peptide was present in the medium during entire 4 h cytotoxicity assay (FIG. 8D). The relative binding affinity of these epitopes to HLA-A2.1 was also measured using an in vitro competitive binding assay (Table 13). G9154 had an higher binding affinity (50% inhibition of the standard peptide at 11 nM) to the HLA-A2.1 molecule than G10154 (1010 nM) which contains an extra leucine at the C-terminus of G9154, and could sensitize T2 cells at lower concentrations than G10154 (FIG. 8A). G9209 also bound to HLA-A2.1 with higher affinity (84 nM) than G10208 (2080 nM) which contains an extra threonine at the N-terminus, and could sensitize T2 cells at lower concentrations of peptide than G10208 (FIG. 8B). Thus, the 9-mer peptides were superior to the corresponding 10 mer peptides in the sensitization of T2 cells to CTL lysis, and they also had higher binding affinities to HLA-A2.1. This was also the case for the identified MART-1 9 and 10 amino acid peptides (M9-2, M10-3, M10-4) (Example 2; Kawakami, Y., et al., (1994). J. Exp. Med. 180:347). The results of the peptide titration in the T2 cell lysis assay correlated with the results of the HLA-A2.1 binding affinity as measured by the in vitro binding assay. The other gp100 epitopes, G9280, G10-4, or G10-5 had binding affinities for HLA-A2.1 with 50 inhibition at 95 nM, 483 nM, or 13 nM, respectively. The HLA-A2.1 binding affinities of the previously identified HLA-A2 restricted melanoma epitopes in MART-1 (Example 2; Kawakami, Y., et al., (1994) J. Exp. Med. 180:347) and tyrosinase (Wolfel, T., (1994) Eur. J. Immunol. 24:759) were also measured (M9-2(397 nM), M10-3(2272 nM), M10-4(5555 nM), T9309(333 nM), T9369 (40 nM)). Except for the 10mer peptides (G10154, G10208, GM-10-3, GM10-4), for which overlapping 9-mer epitopes (G9154, G9209, M9-2) existed, all melanoma epitopes had either high (G9154, G10-5, T9369) or intermediate (G9209, G9280, G10-4, M9-2, T91) binding affinities to HLA-A2.1.
  • Discussion
  • Multiple epitopes in the gp100 human melanoma antigen recognized by 4 TIL which were associated with tumor regression when adoptively transferred to the autologous patients have been identified in this study. Among the 5 epitopes described in this study, G9154 or G10154 appeared to be the most commonly recognized, since these were recognized by 3 of 4 gp100 reactive TIL derived from different patients. Although the G9280 peptide was reported to be recognized by all 5 CTL derived from PBL of different patients (Cox, A. L., et al., (1994)). Science 264:716), it was only recognized by 2 of 4 gp100 reactive TIL in this study. This difference may be due to the sources of T-cells (TIL vs PBL) used.
  • It will be appreciated that the MART-1 peptide M9-2 may also be designated M927, the MART-1 peptide M10-3 may also be designated M1026, and the MART-1 peptide M10-4 may also be designated M1027. It will also be appreciated that the gp100 peptide G10-4 may also be designated G10457 and the gp100 peptide G10-5 may also be designated G10476.
  • TABLE 11
    Summary of antigen recognition by HLA-A2 restricted melanoma specific TIL
    TIL
    1200 620 660 1143 1074 1088 1235 1318 1363 1399
    Clinical* PR PR PR PR NR NR NR NR NR NR
    Response
    Antigen(epitope)
    gp100 + + + +
    (G9154) (G9209) (G9280) (G9280)
    (G10-4) (G9154) (G9154)
    (G10-5)
    MART-1 + + + + + + + + +
    (M9-2) (M9-2) (M9-2) (M9-2) (M9-2) (M9-2) (M9-2) (M9-2) (M9-2)
    tyrosinase
    gp75
    G9(10): gp100-9(10)-mer peptides, M9-2: MART-127-35 peptide Recognition of gp100 by TIL is significantly (p < 0.001)correlated with clinical response for adoptive immunotherapy with HLA-A2 restricted TIL.
    *PR, partial response (>50% reduction in the sum of the product of perpendicular tumor diameters); NR, no response (<50% reduction)
  • TABLE 12
    Recognition of gp100 peptides by TIL
    Target TIL
    cells Peptide (% specific lysis at E:T = 40:1)
    Exp. 1 620-1 620-2 660-1 1143 1200 1235
    624mel none 32 36 47 20 77 11
    397mel none 2 3 0 0 0 0
    T2 none 0 5 3 1 0 2
    T2 M9-2 19 84 69 49 1 86
    T2 G9154 0 21 4 0 100 0
    T2 G10154 3 19 7 4 75 2
    T2 G9209 45 21 0 3 0 0
    T2 G10208 42 36 7 4 2 3
    T2 G9280 2 7 43 11 0 0
    T2 G10-4 0 7 6 0 15 0
    T2 G10-5 2 7 2 1 7 0
    Exp. 2 620-1 620-2 660-2 1143 1200 1235
    624mel none 60 65 74 49 82 18
    397mel none 2 6 0 0 0 0
    T2 none 1 12 1 0 1 2
    T2 M9-2 36 85 50 39 0 60
    T2 G9154 5 27 32 1 78 5
    T2 G10154 4 31 30 2 85 3
    T2 G9209 22 74 5 4 1 3
    T2 G10208 35 80 7 10 1 5
    T2 G9280* 2 9 75 34 1 2
    Exp. 3 660-3 1143 1200 1235
    624mel none 52 15 66 40
    397mel none 5 3 7 4
    T2 none 7 3 7 4
    T2 M9-2 50 62 4 94
    T2 G9280 99 37 9 5
    T2 G10-4 0 0 50 0
    T2 G10-5 14 2 6 5
    Lysis by TIL of T2 cells preincubated with MART1 epitope, M9-2 (AAGIGILTV) and gp100 epitopes, G9154 (KTWGQYWQV), G10154 (KTWGQYWQVL), G9209(ITDQVPFSV), G10208 (TITDQVPFSV), G9280 (YLEPGPVTA), G10-4(LLDGTATLRL), G10-5 (VLYRYGSFSV) at 1 ug/ml (* 1 ng/ml), was measured by 4 h-51Cr release assays. TIL620-1, -2 or TIL660-1, -2, -3 were grown from the same TIL which was administered into the autologous patient, but were separately cultured. 624mel, HLA-A2+ gp100+, MART-1+ melanoma cell line, 397mel, HLA-A2− melanoma cell line. T2 cells, HLA-A2+ T cell-B-cell hybridoma.
    Bold: statistically significant lysis
  • TABLE 13
    The relative binding affinity of the human
    melanoma epitopes to HLA-A2.1
    Peptide Sequence 50% Ratio
    Protein (nM)a Standard.b inhibition to
    gp100 G9154 KTWGQYWQV 11 0.45
    G10154 KTWGQYWQVL 1010 0.005
    G9209 ITDQVPFSV 84 0.06
    G10208 TITDQVPFSV 2080 0.0024
    G9280 YLEPGPVTA 95 0.053
    G10-4 LLDGTATLRL 483 0.01
    G10-5 VLYRYGSFSV 13 0.38
    MART-1 M9-2 AAGIGILTV 395 0.013
    M10-3 EAAGIGILTV 2272 0.0022
    M10-4 AAGIGILTVI 5555 0.0009
    Tyrosinase T91 MLLAVLYCL 333 0.015
    T9369 YMNGTMSQV 40 0.13
    aConcentration of sample peptide required for 50% inhibition of the standard radiolabeled peptide HBC18-27.
    bRatio of the binding affinity of the sample peptide to that of the standard peptide (50% inhibition at 5 nM). Peptides are defined as high (50% inhibition at <50 nM, ratio >0.1), intermediate (50-500 nM, ratio 0.1-0.01) and weak (>500 nM, ratio <0.01) binding peptides.
  • EXAMPLE 5 Modification of melanoma Epitopes for Improvement of Immunogenicity Material and Methods
  • Peptide synthesis and HLA-A2.1 binding assay. Peptides were synthesized by a solid phase method using a multiple peptide synthesizer and purified by HPLC, as previously described (Rivoltini, L et al. (1995) Journal of Immunology Volume 154:2257-2265). The relative binding of peptides to HLA-A2.1, based on the inhibition of binding of a radiolabeled standard peptide to detergent-solubilized MHC molecules, was performed as previously described (Rivoltini, L et al. (1995) Journal of Immunology Volume 154:2257-2265). Briefly, various doses of the test peptides (ranging from 100 μM to 1 nM) were coincubated together with the 5 nM radiolabeled Hbc 18-27 (FLPSDYFPSV) peptide and HLA-A2.1 heavy chain and β2-microglobulin for 2 days at room temperature in the presence of protease inhibitors. The percentage of MHC-bound radioactivity was determined by gel filtration and the 50% inhibitory dose was calculated for each peptide.
  • Induction of peptide specific CTL PBMC were separated from peripheral blood of HLA-A2+ melanoma patients and normal donors by centrifugation on Ficoll-Hypaque gradients and used as fresh or cryopreserved samples. Peptide specific CTL lines were generated as follows: at day 0, PBMC were plated at a concentration of 1.5×106/ml in 24-well plates (2 ml/well) in Iscove's medium containing 10% human AB serum, L-glutamine, antibiotics (CM) and in the presence of 1 ug/ml peptide. Two days later, 12 IU/ml interleukin 2 (IL-2) (Chiron Co., Emeryville, Calif.) were added to the cultures. Lymphocytes were then restimulated weekly as follows: responder cells were harvested, washed once and replated in 24-well plates at a concentration of 2.5×105 cells/ml in CM. Autologous PBMC were thawed, washed twice in PBS, resuspended at 5-8×106 cells/ml in CM and pulsed with 1 ug/ml peptide in 15-ml conical tubes (5 ml/tube) for 3 hours at 37° C. These PBMC (stimulators) were then irradiated at 3000 rads, washed once in PBS and added to the responder cells at responder:stimulator ratios ranging between 1:3 and 1:10. The next day, 12 IU/ml IL-2 were added to the cultures. The activity of these CTL was tested by cytotoxicity assays after at least 2 rounds (14 days) of peptide stimulation. To generate CTL from TIL cultures, the dissociated tumor suspension were cultured for 1-2 days in 10% FCS RPMI-1640 medium to allow tumor cell adherence. The lymphocytes, recovered from the non-adherent fraction, were used for the induction of peptide specific CTL as described above.
  • Assessment of antigen recognition by CTL. 51Cr release cytotoxic assays were performed to detect the recognition of peptide and melanoma cells by CTL. To analyze peptide recognition, T2 cell lines were preincubated for 2 h at 37° C. with 1 ug/ml peptide, washed and used as target cells in a 51Cr release cytotoxic assay. The melanoma lines 624mel was established in our laboratory (See Example 1).
  • In order to make more immunogenic peptides for induction of anti-melanoma T-cells than natural melanoma epitopes, a variety of peptides in which at least 1 amino acid was changed based on consensus motifs in peptides binding to a specific MHC Class I allele Falk, et al. (1991) Nature 351:290; Kubo et al. (1994) J. Immunol 152:3913; Parker, K. et al. (1992) Journal of Immunology 149:3580; Ruppert, J. et al. (1993) Cell 74:929-937) (Tables 14, 15, 16, and 17). Although most of the previously isolated viral epitopes and the naturally processed HLA-A2.1 binding peptides contained leucine or methionine at the 2nd major anchor position and valine at the last major anchor position (dominant anchor amino acids) and had high binding affinity to HLA-A2.1, the isolated MART-1 or gp100 melanoma epitopes contain non-dominant amino acid at major anchor position such as alanine (the 2nd position of M9-2, the 9th position of G9-280) and threonine (the 2nd position of G9-154 and G9-209). The M9-2, G9-209 and G9-280 are not high affinity binders. By changing amino acid at the 1st, 2nd, 3rd or 9th positions which are important for HLA-A2 binding to the peptide, but less important for recognition by T-cell receptors, artificial peptides which can bind to HLA-A2.1 with higher affinity and still be recognized by natural epitope specific T-cells may be generated.
  • Among modified M9-2, G9-280, G9-209, G9-154 peptides, M9-2-2L, M9-2-1F, M9-2-3Y, G9-280-9V, G9-280-9L, G9-280-9I, G9-280-1F, G9-209-2L, G9-209-2M, G9-209-2I, G9-209-1F, G9-209-1Y, G9-209-1W2L, G9-209-1F2L, G9-209-1Y2L have higher binding affinity and were recognized by the original melanoma reactive T-cells. (Tables 14, 15, 16 and 17) PBL stimulated with autologous PBMC pulsed with G9-154-2I, G9-209-1F2L, or G9-280-9V (Tables 18, 19 and 20) recognized and lysed not only the original epitopes but also melanoma tumor cells (624mel) better than PBL stimulated with natural epitopes (G9-154, G9-209, G9-280).
  • These results demonstrated that modified peptides could be used for induction of anti-tumor T-cells instead of natural epitopes. Other peptides which were not recognized by the particular T-cells used in our study, but have higher binding affinity to HLA-A2.1 may induce a different set of T-cells capable of recognizing the original melanoma epitopes in in vitro or in vivo. These modified peptides may be used for induction of anti-melanoma T-cells in vitro and immunization of patients for the treatment of patients with melanoma or for the prevention of melanoma.
  • TABLE 14
    Modified MART-1 M9-2 peptides
    Binding Recognition
    affinity by M9-2
    to HLA-A2.1 reactive
    Peptide Sequence (nM) T-cell
    M9-2 parent AAGIGILTV 1064 +
    M9-2-2L ALGIGILTV 10 +
    M9-2-2M AMGIGILTV 14
    M9-2-2I AIGIGILTV 77
    M9-2-1W WAGIGILTV 1351 +
    M9-2-1F FAGIGILTV 244 +
    M9-2-1Y YAGIGILTV 136
    M9-2-3W AAWIGILTV 65
    M9-2-3F AAFIGILTV 67
    M9-2-3Y AAYIGILTV 102 +
    M9-2-1K2L KLGIGILTV 14
    M9-2-1K2M KMGIGILTV 27
    M9-2-1K2I KIGIGILTV 94
    M9-2-1W2L WLGIGILTV 11
    M9-2-1F2L FLGIGILTV 1.8
    M9-2-1Y2L YLGIGILTV 3.2
    M9-2-2L3W ALWIGILTV 5.5
    M9-2-2L3F ALFIGILTV 1.4
    M9-2-2L3Y ALYIGILTV 3.7
  • TABLE 15
    Modified gp100 G9-154 peptides
    Binding Recognition
    affinity by G9-154
    to HLA-A2.1 reactive
    Peptide Sequence (nM) T-cell
    G9-154 parent KTWGQYWQV 5.7 +
    G9-154-2L KLWGQYWQV 2 +
    G9-154-2M KMWGQYWQV 6.5 +
    G9-154-2I KIWGQYWQV 3 +
    09-154-1W WTWGQYWQV 60
    09-154-1F FTWGQYWQV 1.6
    G9-154-1Y YTWGQYWQV 2.5
    09-154-1A ATWGQYWQV 5.2 +
    G9-154-1L LTWGQYWQV 3.4 +
    G9-154-3Y KTYGQYWQV 30 +
    09-154-3F KTFGQYWQV 21 +
    G9-154-1A2L ALWGQYWQV 2.3 +
    G9-154-1L2L LLWGQYWQV 1.6 +
    G9-154-1W2L WLWGQYWQV 2.8
    09-154-1F2L FLWGQYWQV 2.6
    G9-154-1Y2L YLWGQYWQV 1.7
  • TABLE 16
    Modified gp100 G9-209 peptides
    Binding Recognition
    affinity by G9-209
    to HLA-A2.1 reactive
    Peptide Sequence (nM)* T-cell
    G9-209 parent ITDQVPFSV 172 +
    G9-209-2L ILDQVPFSV 3.3 +
    G9-209-2M IMDQVPFSV 19 +
    G9-209-2I IIDQVPFSV 40 +
    G9-209-1F FTDQVPFSV 61 +
    G9-209-1W WTDQVPFSV 711 +
    G9-209-1Y YTDQVPFSV 85 +
    G9-209-3W ITWQVPFSV 34
    G9-209-3F ITFQVPFSV 66
    G9-209-3Y ITYQVPFSV 33
    G9-209-3A ITAQVPFSV 95
    G9-209-3M ITMQVPFSV 40
    G9-209-3S ITSQVPFSV 649
    G9-209-2L3W ILWQVPFSV 1.7
    G9-209-2L3F ILFQVPFSV 2
    G9-209-2L3Y ILYQVPFSV 5
    G9-209-2L3A ILAQVPFSV 11
    G9-209-2L3M ILMQVPFSV 7.6
    G9-209-2L3S ILSQVPFSV 20
    G9-209-1W2L WLDQVPFSV 12 +
    G9-209-1F2L FLDQVPFSV 2.2 +
    G9-209-1Y2L YLDQVPFSV 2.3 +
    *a Concentration of sample peptide required for 50% inhibition of the standard radiolabeled peptide HBC18-27. Peptides are defined as high (50% inhibition at <50 nM), intermediate (50-500 nM) and weak (>500 nM) binding peptides. (see ref gp100 epitope)
  • TABLE 17
    Modified gp100 G9-280 peptides
    Binding Recognition
    affinity by G9-280
    to HLA-A2.1 reactive
    Peptide Sequence (nM) T-cell
    G9-280 parent YLEPGPVTA 455 +
    G9-280-9V YLEPGPVTV 48 +
    G9-280-9L YLEPGPVTL 88 +
    G9-280-9I YLEPGPVTI 65 +
    G9-280-1F FLEPGPVTA 125 +
    G9-280-1W WLEPGPVTA 833 +
    G9-280-3Y YLYPGPVTA 17
    G9-280-3W YLWPGPVTA 3.2
    G9-280-3F YLFPGPVTA 3.2
    G9-280-3M YLMPGPVTA 4.3
    G9-280-3S YLSPGPVTA 42
    G9-280-3A YLAPGPVTA 9.3
    G9-280-3M9V YLMPGPVTV 12
    G9-280-3S9V YLSPGPVTV 23
    G9-280-3A9V YLAPGPVTV 15
    G9-280-3Y9V YLYPGPVTV 8.9
    G9-280-3F9V YLFPGPVTV 5.8
    G9-280-3W9V YLWPGPVTV 7.4
  • TABLE 18
    Induction of anti-melanoma CTL using modified
    G9-154 peptide
    Effector T-cells
    PBL stimulated PBL stimulated
    with G9-154 with G9-154-2I
    Target % specific lysis (E:T = 3D40:1)
    T2 11 1
    T2+G9-154 14 37
    T2+G9-154-2I 8 38
    624mel 5 23
    51Cr release assay was performed after 4 times stimulation with autologous PBMC preincubated with peptides.
  • TABLE 19
    Induction of anti-melanoma CTL using modified
    G9-209 peptide
    Effector T-cells
    PBL stimulated PBL stimulated
    with G9-209 with G9-209-1F2L
    Target % specific lysis (E:T = 3D40:1)
    T2 0 0
    T2+G9-209 6 85
    T2+G9-209-1F2L 1 86
    624mel 4 63
    51Cr release assay was performed after 4 times stimulation with autologous PBMC preincubated with peptides.
  • TABLE 20
    Induction of anti-melanoma CTL using modified
    G9-280 peptide
    Effectoe T-cells
    PBL stimulated PBL stimulated
    with G9-280 with G9-280-9V
    Target % specific lysis (E:T = 3D40:1)
    T2 3 0
    T2+G9-280 11 87
    T2+G9-280-9V 8 58
    624mel 11 71
    51Cr release assay was performed after 4 times stimulation with autologous PBMC preincubated with peptides.
  • EXAMPLE 6 MART-1 vaccines as a Treatment for Melanoma in Mammals
  • MART-1 vaccines may be efficacious in treating mammals afflicted with melanoma. For example, MART-1 vaccines may be administered to individuals. Mammals can be immunized with the MART-1 proteins, peptides or modified peptides described herein in ranges of about 1 mg-to to about 100 mg. Alternatively mammals, preferably humans may be immunized with the MART-1 nucleic acid sequence inserted into a viral vector such as vaccinia virus, adenovirus or fowl pox virus. A range of about 106 to about 1011 viral particles carrying the MART-1 nucleic acid sequences corresponding to immunogenic MART-1 peptides or modified peptides or analogs therof, may be administered per mammal, preferably a human. The mammals will be monitored for antibodies to the immunogen or increase in cytotoxic lymphocytes (CTL) recognizing the immunogen by conventional methods or alleviation of clinical signs and symptoms of the active disease. Specific parameters to be assessed include production of immune cells that recognize the vaccine antigen or tumor regression. Such vaccines may be administered either prophylactically or therapeutically. Mammals may also be immunized with the gp-100 nucleic acid sequence inserted into a retroviral vector or GP-100 immunogenic peptides or modified peptides or analogs thereof. Suggested dose ranges of the antigen in retroviruses that may be used are about 106 to about 1011 viral particles per mammal, preferably a human. Response and efficacy of the retroviral vaccines will be assessed as described above.
  • EXAMPLE 7 Use of Lymphocytes Sensitized to Immunogenic Peptides Derived from Melanoma Antigens for Therapeutically Treating Mammals Afflicted with Melanoma
  • T-lymphocytes presensitized to the melanoma antigen may be effective in therapeutically treating mammals afflicted with melanoma. The T-lymphocytes will be isolated from peripheral blood lymphocytes or tumor infiltrating lymphocytes and exposed in vitro to the MART-1 protein or peptide. T-lymphocytes are isolated from peripheral blood or melanoma tumor suspensions and cultured in vitro (Kawakami, Y. et al. (1988) J. Exp. Med. 168: 2183-2191). The T-lymphocytes are exposed to the MART-1 peptide AAGIGILTV for a period of about to 1-16 hours at a concentration of about 1 to about 10 mg/ml. T-lymphocytes exposed to the antigen will be administered to the mammal, preferably a human at about 109 to about 1012 lymphocytes per mammal. Alternatively, the T-lymphocytes may be exposed to the modified MART-1 peptides. The lymphocytes may be administered either intravenously, intraperitoneally or intralesionally. This treatment may be administered concurrently with other therapeutic treatments such as cytokines, radiotherapy, surgical excision of melanoma lesions and chemotherapeutic drugs, adoptive T lymphocyte therapy. Alternatively, the T-lymphocytes may be exposed to the gp100 immunogenic peptides or modified immunogenic peptides described herein.
  • The present invention is not to be limited in scope by the nucleic acid sequences deposited, since the deposited embodiments is intended as a single illustration of one aspect of the invention and any sequences which are functionally equivalent are within the scope of this invention. Indeed, various modifications of the invention in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description and accompanying drawings. Such modifications are intended to fall within the scope of the dependent claims.

Claims (16)

1.-66. (canceled)
67. An isolated host cell transformed or transfected with the recombinant expression vector comprising all or part of a nucleic acid encoding a MART-1 protein (SEQ ID NO: 2) in a manner to allow expression of said protein encoded by said recombinant expression vector.
68. A vector comprising a nucleic acid encoding an immunogenic peptide comprising 5-20 contiguous amino acids of gp100 (SEQ ID NO: 27) with at least one amino acid modification in the 5-20 contiguous amino acids of SEQ ID NO: 27, wherein the immunogenic peptide reacts with T-lymphocytes.
69. The vector of claim 68, wherein said modification an amino acid substitution.
70. The vector of claim 69, wherein the amino acid substitution is located at an amino acid position of the immunogenic peptide selected from the group consisting of: (i) the first position, (ii) the second position, (iii) the third position, (iv) the ninth position, (v) tenth position, and (vi) combinations of at least two of (i)-(v).
71. The vector of claim 70, wherein the immunogenic peptide is selected from the group consisting of a group consisting of KTWGOYWQV (SEQ ID NO: 46) with at least one amino acid modification in SEQ ID NO: 46, KTWGQYWQVL (SEQ ID NO: 47) with at least one amino acid modification in SEQ ID NO: 47, ITDQVPFSV (SEQ ID NO: 48) with at least one amino acid modification in SEQ ID NO: 48, TITDQVPFSV (SEQ ID NO: 49) with at least one amino acid modification in SEQ ID NO: 49, LLDGTATLRL (SEQ ID NO: 33) with at least one amino acid modification in SEQ ID NO: 33, VLYRYGSFSV (SEQ ID NO: 34) with at least one amino acid modification in SEQ ID NO: 34, VLKRCLLHL (SEQ ID NO: 36) with at least one amino acid modification in SEQ ID NO: 36, ALDGGNKHFL (SEQ ID NO: 35) with at least one amino acid modification in SEQ ID NO: 35, VLPSPACQLV (SEQ ID NO: 37) with at least one amino acid modification in SEQ ID NO: 37, YLEPGPVTA (SEQ ID NO: 40) with at least one amino acid modification in SEQ ID NO: 40, SLADTNSLAV (SEQ ID NO: 38) with at least one amino acid modification in SEQ ID NO: 38, SVSVSQLRA (SEQ ID NO: 39) with at least one amino acid modification in SEQ ID NO: 39, and LNVSLADTN (SEQ ID NO: 41) with at least one amino acid modification in SEQ ID NO: 41.
72. A vector comprising an isolated nucleic acid encoding an immunogenic peptide comprising an amino acid sequence of the formula selected from the group consisting of X1X2X3 GQYWQX4, X1X2X3QVPFSX4 and X1X2X3PGPVTX4, wherein: X1 is any naturally occurring amino acid; X2 is any hydrophobic aliphatic amino acid; X3 is any naturally occurring amino acid; and X4 is any hydrophobic aliphatic amino acid.
73. The vector of claim 72, wherein X1 is selected from the group consisting of methionine, leucine, alanine, glycine, threonine, isoleucine, valine, tyrosine, serine, tryptophan, phenylalanine, lysine and aspartic acid.
74. The vector of claim 72, wherein X2 is selected from the group consisting of methionine, leucine, alanine, glycine, isoleucine, valine and threonine.
75. The vector of claim 72, wherein X3 is selected from the group consisting of methionine, leucine, alanine, glycine, threonine, isoleucine, tyrosine, valine, tryptophan, phenylalanine, serine, lysine and aspartic acid.
76. The vector of claim 72, wherein X4 is selected from the group consisting of methionine, leucine, alanine, glycine, isoleucine, valine and threonine.
77. The vector of claim 72, wherein the immunogenic peptide is selected from the group consisting of KTWGQYWQV (SEQ ID NO: 46), KLWGQYWQV (SEQ ID NO: 68), KMQGQYWQV (SEQ ID NO: 69), KIWGQYWQV (SEQ ID NO: 70), WTWGQYWQV (SEQ ID NO: 71), FTWGQYWQV (SEQ ID NO: 72), YTWGQYWQV (SEQ ID NO: 73), ATWGQYWQV (SEQ ID NO: 74), LTWGQYWQV (SEQ ID NO: 75), KTYGQYWQV (SEQ ID NO: 76), KTFGQYWQV (SEQ ID NO: 77), ALWGQYWQV (SEQ ID NO: 78), LLWGQYWQV (SEQ ID NO: 79), WLWGQYWQV (SEQ ID NO: 80), FLWGQYWQV (SEQ ID NO: 81), and YLWGQYWQV (SEQ ID NO: 82).
78. The vector of claim 72, wherein the immunogenic peptide is selected from the group consisting of ITDQVPFSV (SEQ ID NO: 48), ILDQVPFSV (SEQ ID NO: 83), IMDQVPFSV (SEQ ID NO: 84), IIDQVPFSV (SEQ ID NO: 85), FTDQVPFSV (SEQ ID NO: 86), WTDQVPFSV (SEQ ID NO: 87), YTDQVPFSV (SEQ ID NO: 88), ITWQVPFSV (SEQ ID NO: 89), ITFQVPFSV (SEQ ID NO: 90), ITYQVPFSV (SEQ ID NO: 91), ITAQVPFSV (SEQ ID NO: 92), ITMQVPFSV (SEQ ID NO: 93), ITSQVPFSV (SEQ ID NO: 94), ILWQVPFSV (SEQ ID NO: 95), ILFQVPFSV (SEQ ID NO: 96), ILYQVPFSV (SEQ ID NO: 97), ILAQVPFSV (SEQ ID NO: 98), ILMQVPFSV (SEQ ID NO: 99), ILSQVPFSV (SEQ ID NO: 100), WLDQVPFSV (SEQ ID NO: 101), FLDQVPFSV (SEQ ID NO: 102), and YLDQVPFSV (SEQ ID NO: 103).
79. The vector of claim 72, wherein the immunogenic peptide is selected from the group consisting of YLEPGPVTA (SEQ ID NO. 40), YLEPGPVTV (SEQ ID NO:104), YLEPGPVTL (SEQ ID NO:105), YLEPGPVTI (SEQ ID NO:106), FLEPGPVTA (SEQ ID NO: 107), WLEPGPVTA (SEQ ID NO: 108), YLYPGPVTA (SEQ ID NO: 109), YLWPGPVTA (SEQ ID NO: 110), YLFPGPVTA (SEQ ID NO: 111), YLMPGPVTA (SEQ ID NO: 112), YLSPGPVTA (SEQ ID NO: 113), YLAPGPVTA (SEQ ID NO: 114), YLMPGPVTV (SEQ ID NO: 115), YLSPGPVTV (SEQ ID NO: 116), YLAPGPVTV (SEQ ID NO: 117), YLYPGPVTV (SEQ ID NO: 118), YLFPGPVTV (SEQ ID NO: 119), and YLWPGPVTV (SEQ ID NO: 120).
80. A host cell comprising the vector of claim 68.
81. A host cell comprising the vector of claim 72.
US11/932,255 1994-04-22 2007-10-31 Melanoma antigens and their use in diagnostic and therapeutic methods Expired - Fee Related US7803614B2 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US11/932,255 US7803614B2 (en) 1994-04-22 2007-10-31 Melanoma antigens and their use in diagnostic and therapeutic methods
US12/851,729 US8030280B2 (en) 1994-04-22 2010-08-06 Melanoma antigens and their use in diagnostic and therapeutic methods
US13/192,804 US8273724B2 (en) 1994-04-22 2011-07-28 Melanoma antigens and their use in diagnostic and therapeutic methods

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
US08/231,565 US5874560A (en) 1994-04-22 1994-04-22 Melanoma antigens and their use in diagnostic and therapeutic methods
US08/417,174 US5844075A (en) 1994-04-22 1995-04-05 Melanoma antigens and their use in diagnostic and therapeutic methods
US09/073,138 US6537560B1 (en) 1994-04-22 1998-05-05 Melanoma antigens and their use in diagnostic and therapeutic methods
US09/267,439 US6270778B1 (en) 1994-04-22 1999-03-12 Melanoma antigens and their use in diagnostic and therapeutic methods
US09/898,860 US6965017B2 (en) 1994-04-22 2001-07-03 Melanoma antigens and their use in diagnostic and therapeutic methods
US10/685,977 US7232887B2 (en) 1994-04-22 2003-10-15 Melanoma antigens and their use in diagnostic and therapeutic methods
US11/751,233 US7612044B2 (en) 1994-04-22 2007-05-21 Melanoma antigens and their use in diagnostic and therapeutic methods
US11/772,277 US7807805B2 (en) 1994-04-22 2007-07-02 Melanoma antigens and their use in diagnostic and therapeutic methods
US11/932,255 US7803614B2 (en) 1994-04-22 2007-10-31 Melanoma antigens and their use in diagnostic and therapeutic methods

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/772,277 Continuation US7807805B2 (en) 1994-04-22 2007-07-02 Melanoma antigens and their use in diagnostic and therapeutic methods

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/851,729 Continuation US8030280B2 (en) 1994-04-22 2010-08-06 Melanoma antigens and their use in diagnostic and therapeutic methods

Publications (2)

Publication Number Publication Date
US20080233637A1 true US20080233637A1 (en) 2008-09-25
US7803614B2 US7803614B2 (en) 2010-09-28

Family

ID=22869773

Family Applications (6)

Application Number Title Priority Date Filing Date
US08/231,565 Expired - Lifetime US5874560A (en) 1994-04-22 1994-04-22 Melanoma antigens and their use in diagnostic and therapeutic methods
US08/417,174 Expired - Lifetime US5844075A (en) 1994-04-22 1995-04-05 Melanoma antigens and their use in diagnostic and therapeutic methods
US09/007,961 Expired - Lifetime US5994523A (en) 1994-04-22 1998-01-16 Melanoma antigens and their use in diagnostic and therapeutic methods
US11/932,255 Expired - Fee Related US7803614B2 (en) 1994-04-22 2007-10-31 Melanoma antigens and their use in diagnostic and therapeutic methods
US12/851,729 Expired - Fee Related US8030280B2 (en) 1994-04-22 2010-08-06 Melanoma antigens and their use in diagnostic and therapeutic methods
US13/192,804 Expired - Fee Related US8273724B2 (en) 1994-04-22 2011-07-28 Melanoma antigens and their use in diagnostic and therapeutic methods

Family Applications Before (3)

Application Number Title Priority Date Filing Date
US08/231,565 Expired - Lifetime US5874560A (en) 1994-04-22 1994-04-22 Melanoma antigens and their use in diagnostic and therapeutic methods
US08/417,174 Expired - Lifetime US5844075A (en) 1994-04-22 1995-04-05 Melanoma antigens and their use in diagnostic and therapeutic methods
US09/007,961 Expired - Lifetime US5994523A (en) 1994-04-22 1998-01-16 Melanoma antigens and their use in diagnostic and therapeutic methods

Family Applications After (2)

Application Number Title Priority Date Filing Date
US12/851,729 Expired - Fee Related US8030280B2 (en) 1994-04-22 2010-08-06 Melanoma antigens and their use in diagnostic and therapeutic methods
US13/192,804 Expired - Fee Related US8273724B2 (en) 1994-04-22 2011-07-28 Melanoma antigens and their use in diagnostic and therapeutic methods

Country Status (5)

Country Link
US (6) US5874560A (en)
EP (1) EP2311952A3 (en)
JP (1) JP2010178739A (en)
CA (1) CA2722364C (en)
FI (1) FI123050B (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8030280B2 (en) 1994-04-22 2011-10-04 The United States Of America As Represented By The Secretary, Department Of Health And Human Services Melanoma antigens and their use in diagnostic and therapeutic methods

Families Citing this family (113)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5856091A (en) * 1993-03-18 1999-01-05 Ludwig Institute For Cancer Research Isolated nucleic acid sequence coding for a tumor rejection antigen precursor processed to at least one tumor rejection antigen presented by HLA-A2
US6660276B1 (en) 1994-02-16 2003-12-09 The University Of Virginia Patent Foundation Peptides recognized by melanoma-specific cytotoxic lymphocytes, and uses therefor
DK0668350T4 (en) * 1994-02-16 2009-02-23 Us Gov Health & Human Serv Melanoma-associated antigen, epitopes thereof and melanoma vaccines
AU706443B2 (en) * 1994-04-22 1999-06-17 Government Of The United States Of America, As Represented By The Secretary Of The Department Of Health And Human Services, The Melanoma antigens
ES2203782T3 (en) * 1996-01-17 2004-04-16 Imperial College Innovations Limited IMMUNOTHERAPY THAT USES CYTOTOXIC T LYMPHOCYTES (CTL).
US7015312B1 (en) * 1996-02-09 2006-03-21 The United States Of America As Represented By The Department Of Health And Human Services Antibodies to the protein product encoded by ORF3 of the TRP-1 gene and compositions and kits thereof
US20040156861A1 (en) * 1996-07-11 2004-08-12 Figdor Carl Gustav Melanoma associated peptide analogues and vaccines against melanoma
US20020164318A1 (en) * 1997-02-18 2002-11-07 Houghton Alan N. Cell lines and vectors for stimulation of an immune response to a differentiation antigen stimulated by an altered antigen
US6326200B1 (en) * 1997-06-23 2001-12-04 Ludwig Institute For Cancer Research Isolated nona-and decapeptides which bind to HLA molecules, and the use thereof
US6025470A (en) * 1997-06-23 2000-02-15 Ludwig Institute For Cancer Research Isolated nona- and decapeptides which bind to HLA molecules, and the use thereof
US6872518B2 (en) * 1997-09-22 2005-03-29 University Of Rochester Methods for selecting polynucleotides encoding T cell epitopes
AU1628599A (en) * 1997-12-05 1999-06-28 Medical College Of Georgia Research Institute, Inc. Regulation of t cell-mediated immunity by tryptophan
US20030138808A1 (en) * 1998-02-19 2003-07-24 Simard John J.L. Expression vectors encoding epitopes of target-associated antigens
EP1071325A4 (en) * 1998-03-20 2004-12-22 Genzyme Corp Compositions and methods for antigen-specific vaccination
US6465180B1 (en) * 1999-03-17 2002-10-15 The Regents Of The University Of California Detection of premalignant melanocytes
GB9908263D0 (en) * 1999-04-13 1999-06-02 Binding Site The Limited Eliciting improved immune responses
CA2382774A1 (en) * 1999-05-25 2000-11-30 Human Genome Sciences, Inc. Meth1 and meth2 polynucleotides and polypeptides
AU7917200A (en) * 1999-09-30 2001-04-30 Institut Pasteur Hybrid or chimeric polynucleotides, proteins, and compositions comprising hepatitis b virus sequences
GB9924351D0 (en) 1999-10-14 1999-12-15 Brennan Frank Immunomodulation methods and compositions
DE60038971D1 (en) 1999-10-22 2008-07-03 Aventis Pasteur PROCESS FOR EXPLORING AND / OR REINFORCING THE IMMUNE RESPONSE AGAINST TUMORANTIGENE
WO2001070767A2 (en) * 2000-03-20 2001-09-27 Genzyme Corporation Therapeutic anti-melanoma compounds
FR2806727A1 (en) * 2000-03-23 2001-09-28 Pf Medicament MOLECULE OF PHARMACEUTICAL INTEREST COMPRISING IN THE N-TERMINAL END A GLUTAMIC ACID OR GLUTAMINE IN THE FORM OF A PHYSIOLOGICALLY ACCEPTABLE ACID ADDITION SALT
US20030194696A1 (en) * 2000-03-28 2003-10-16 University Of Rochester Methods of producing a library and methods of selecting polynucleotides of interest
US7067251B2 (en) * 2000-03-28 2006-06-27 University Of Rochester Methods of directly selecting cells expressing inserts of interest
US20030215425A1 (en) * 2001-12-07 2003-11-20 Simard John J. L. Epitope synchronization in antigen presenting cells
US6861234B1 (en) 2000-04-28 2005-03-01 Mannkind Corporation Method of epitope discovery
NZ521715A (en) * 2000-04-28 2008-01-31 Mannkind Corp Method of identifying and producing antigen peptides and use thereof as vaccines
EP1282702B1 (en) * 2000-05-10 2006-11-29 Sanofi Pasteur Limited Immunogenic polypeptides encoded by mage minigenes and uses thereof
AU2001264764A1 (en) * 2000-05-31 2001-12-11 Genzyme Corporation Therapeutic anti-melanoma compounds
CA2416890A1 (en) * 2000-08-04 2002-02-14 Genzyme Corporation Therapeutic anti-melanoma compounds
EP1222928A3 (en) * 2001-01-16 2003-11-05 Universität Zürich Institut für Medizinische Virologie Pharmaceutical compositions for treating or preventing cancer, especially melanoma
US20020177547A1 (en) * 2001-01-16 2002-11-28 Karin Molling Pharmaceutical compositions for treating or preventing cancer
EP1359937A4 (en) * 2001-02-14 2004-07-28 Genzyme Corp Altered peptide ligands
ES2643582T3 (en) 2001-02-20 2017-11-23 Janssen Pharmaceuticals, Inc. Artificial Drosophila antigen presenting cell to prepare suspension of CD8 cells for use in cancer treatment
US20040071671A1 (en) * 2001-02-20 2004-04-15 Leturcq Didier J. Cell therapy method for the treatment of tumors
US7723111B2 (en) * 2001-03-09 2010-05-25 The United States Of America As Represented By The Department Of Health And Human Services Activated dual specificity lymphocytes and their methods of use
KR100669065B1 (en) * 2001-04-03 2007-01-15 다카라 바이오 가부시키가이샤 Cytotoxic t lymphocyte
WO2003008537A2 (en) * 2001-04-06 2003-01-30 Mannkind Corporation Epitope sequences
EP1752160A3 (en) 2001-04-06 2007-05-30 Mannkind Corporation Epitope sequences
US20040202654A1 (en) * 2001-04-09 2004-10-14 Mayo Foundation For Medical Education & Research Methods and materials for cancer treatment
EP2687593A1 (en) * 2001-05-15 2014-01-22 Ortho-McNeil Pharmaceutical, Inc. Ex-vivo priming for generating cytotoxic T lymphocytes specific for non-tumor antigens to treat autoimmune and allergic disease
US20080220015A1 (en) * 2001-08-03 2008-09-11 Nokad, S.A. Method of modulating neutralizing antibodies formation in mammals, and uses thereof in gene therapy, animal trangenesis and in functional inactivation of endogenous proteins
US20030113919A1 (en) * 2001-08-17 2003-06-19 Aventis Pasteur, Ltd. Immunogenic targets for melanoma
WO2003018610A2 (en) * 2001-08-22 2003-03-06 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services Peptides of melanoma antigen and their use in diagnostic, prophylactic and therapeutic methods
CN101024842A (en) * 2001-11-07 2007-08-29 曼康公司 Expression vectors encoding epitopes of target-associated antigens and its design method
US6783944B2 (en) 2002-01-04 2004-08-31 The Regents Of The University Of California Genetic changes in atypical nodular proliferations in congenital melanocytic nevi
DE10229872A1 (en) * 2002-07-03 2004-01-29 Curevac Gmbh Immune stimulation through chemically modified RNA
US20040018622A1 (en) * 2002-07-17 2004-01-29 Mitchell Lloyd G. Spliceosome mediated RNA trans-splicing for correction of skin disorders
EP1556082A1 (en) * 2002-10-22 2005-07-27 Aventis Pasteur Limited Anti-cancer vaccines and high-dose cytokines as adjuvants
US7537767B2 (en) * 2003-03-26 2009-05-26 Cytis Biotechnology Ag Melan-A- carrier conjugates
RU2351362C2 (en) * 2003-03-26 2009-04-10 Цитос Байотекнолоджи Аг CONJUGATES OF PEPTIDE Melan-A, VIRUS-LIKE PARTICLE ANALOGUE
US20060210588A1 (en) * 2003-03-26 2006-09-21 Cytos Biotechnology Ag Hiv-peptide-carrier-conjugates
US20050186289A1 (en) * 2003-04-01 2005-08-25 Medical College Of Georgia Research Institute, Inc. Regulation of T cell-mediated immunity by D isomers of inhibitors of indoleamine-2,3-dioxygenase
US7598287B2 (en) * 2003-04-01 2009-10-06 Medical College Of Georgia Research Institute, Inc. Use of inhibitors of indoleamine-2,3-dioxygenase in combination with other therapeutic modalities
US7178491B2 (en) * 2003-06-05 2007-02-20 Caterpillar Inc Control system and method for engine valve actuator
US7811983B2 (en) * 2003-06-11 2010-10-12 The University Of Chicago Increased T-cell tumor infiltration and eradication of metastases by mutant light
ES2297479T3 (en) 2003-06-11 2008-05-01 The University Of Chicago INFILTRATION INCREASED IN TUMOR OF CELLS T BY THE MUTANT LIGTH.
ATE546153T1 (en) * 2003-06-17 2012-03-15 Mannkind Corp COMBINATIONS OF TUMOR-ASSOCIATED ANTIGENS FOR THE TREATMENT OF DIFFERENT TYPES OF CANCER
US7696322B2 (en) * 2003-07-28 2010-04-13 Catalent Pharma Solutions, Inc. Fusion antibodies
EP1670513B1 (en) * 2003-10-06 2013-01-23 Cedars-Sinai Medical Center Cox-2 inhibitors and dendritic cells for use in the treatment of cancer.
US20080038838A1 (en) * 2003-10-10 2008-02-14 Towa Kagaku Co., Ltd. Dioxin-Binding Material and Method of Detecting or Quantifying Dioxin
DE602004018819D1 (en) * 2003-10-29 2009-02-12 Kumamoto Tech & Ind Found Diagnostic procedure for malignant melanoma
US7858323B2 (en) 2004-06-09 2010-12-28 The Regents Of The University Of Michigan Phage microarray profiling of the humoral response to disease
US20060008468A1 (en) * 2004-06-17 2006-01-12 Chih-Sheng Chiang Combinations of tumor-associated antigens in diagnostics for various types of cancers
MXPA06014768A (en) * 2004-06-17 2007-06-22 Mannkind Corp Epitope analogs.
US20060159689A1 (en) * 2004-06-17 2006-07-20 Chih-Sheng Chiang Combinations of tumor-associated antigens in diagnostics for various types of cancers
US20070154889A1 (en) * 2004-06-25 2007-07-05 Veridex, Llc Methods and reagents for the detection of melanoma
US7576323B2 (en) 2004-09-27 2009-08-18 Johns Hopkins University Point-of-care mass spectrometer system
PL1833506T3 (en) 2004-12-29 2016-01-29 Mannkind Corp Use of compositions comprising various tumor-associated antigens as anti-cancer vaccines
EP1835932A2 (en) * 2004-12-29 2007-09-26 Mannkind Corporation Methods to elicit, enhance and sustain immune responses against mhc class i-restricted epitopes, for prophylactic or therapeutic purposes
US20100080792A1 (en) * 2005-06-16 2010-04-01 Dorothee Herlyn Method for Identifying a MHC Class II-Dependent Tumor-Associated T Helper Cell Antigen
NZ587601A (en) 2005-06-17 2012-05-25 Mannkind Corp Multivalent Entrain-and-Amplify Immunotherapeutics for Carcinoma
CA2612516C (en) 2005-06-17 2015-03-24 Mannkind Corporation Methods and compositions to elicit multivalent immune responses against dominant and subdominant epitopes, expressed on cancer cells and tumor stroma
CA2612494A1 (en) * 2005-06-17 2006-12-28 Mannkind Corporation Epitope analogues
CA2523032A1 (en) * 2005-10-07 2007-04-07 Immunovaccine Technologies Inc. Vaccines for cancer therapy
US20070178113A1 (en) * 2005-11-22 2007-08-02 Backstrom B T Superantigen conjugate
DE102006035618A1 (en) * 2006-07-31 2008-02-07 Curevac Gmbh New nucleic acid useful as immuno-stimulating adjuvant for manufacture of a composition for treatment of cancer diseases e.g. colon carcinomas and infectious diseases e.g. influenza and malaria
AU2007280690C1 (en) 2006-07-31 2012-08-23 Curevac Gmbh Nucleic acid of formula (I): GIXmGn, or (II): CIXmCn, in particular as an immune-stimulating agent/adjuvant
US8129184B2 (en) * 2006-09-26 2012-03-06 Cedars-Sinai Medical Center Cancer stem cell antigen vaccines and methods
US8871211B2 (en) * 2006-09-28 2014-10-28 Cedars-Sinai Medical Center Cancer vaccines and vaccination methods
WO2008098183A2 (en) * 2007-02-08 2008-08-14 The University Of Chicago Combination therapy for treating cancer
US8263081B2 (en) 2007-05-14 2012-09-11 The University Of Chicago Antibody-light fusion products for cancer therapeutics
US20090131355A1 (en) * 2007-05-23 2009-05-21 Adrian Ion Bot Multicistronic vectors and methods for their design
WO2009030254A1 (en) 2007-09-04 2009-03-12 Curevac Gmbh Complexes of rna and cationic peptides for transfection and for immunostimulation
US9498493B2 (en) 2007-09-27 2016-11-22 Immunovaccine Technologies Inc. Use of liposomes in a carrier comprising a continuous hydrophobic phase for delivery of polynucleotides in vivo
AU2009210266B2 (en) 2008-01-31 2015-01-29 CureVac SE Nucleic acids comprising formula (NuGlXmGmGnNv)a and derivatives thereof as an immunostimulating agents/adjuvants
EP2296696B1 (en) 2008-06-05 2014-08-27 ImmunoVaccine Technologies Inc. Compositions comprising liposomes, an antigen, a polynucleotide and a carrier comprising a continuous phase of a hydrophobic substance
DK2328923T3 (en) * 2008-09-02 2016-03-21 Cedars Sinai Medical Center CD133 epitopes
WO2010037408A1 (en) 2008-09-30 2010-04-08 Curevac Gmbh Composition comprising a complexed (m)rna and a naked mrna for providing or enhancing an immunostimulatory response in a mammal and uses thereof
AU2009308707A1 (en) * 2008-10-31 2010-05-06 Biogen Idec Ma Inc. LIGHT targeting molecules and uses thereof
US20100310591A1 (en) * 2009-01-28 2010-12-09 Robert Humphreys Ii-KEY HYBRID PEPTIDES THAT MODULATE THE IMMUNE RESPONSE TO INFLUENZA
WO2010129895A2 (en) * 2009-05-07 2010-11-11 Immunocellular Therapeutics, Ltd. Cd133 epitopes
US20110053829A1 (en) 2009-09-03 2011-03-03 Curevac Gmbh Disulfide-linked polyethyleneglycol/peptide conjugates for the transfection of nucleic acids
US20110274723A1 (en) 2009-10-23 2011-11-10 Mannkind Corporation Cancer immunotherapy and method of treatment
EP3450979A3 (en) 2010-03-17 2019-04-24 The Regents of The University of Michigan Using phage epitopes to profile the immune response
WO2011140284A2 (en) 2010-05-04 2011-11-10 Fred Hutchinson Cancer Research Center Conditional superagonist ctl ligands for the promotion of tumor-specific ctl responses
DK2449113T3 (en) 2010-07-30 2016-01-11 Curevac Ag Complex formation of nucleic acids with the disulfide cross-linked cationic components for transfection and immunostimulation
US9040046B2 (en) 2011-01-31 2015-05-26 Kai Xu Sodium pump antibody agonists and methods of treating heart disease using the same
WO2013049941A1 (en) 2011-10-06 2013-04-11 Immunovaccine Technologies Inc. Liposome compositions comprising an adjuvant that activates or increases the activity of tlr2 and uses thereof
SG11201401632WA (en) * 2011-10-20 2014-05-29 California Stem Cell Inc Antigen presenting cancer vaccine
CN104284680A (en) 2011-12-15 2015-01-14 芝加哥大学 Methods and compositions for cancer therapy using mutant light molecules with increased affinity to receptors
WO2013113326A1 (en) 2012-01-31 2013-08-08 Curevac Gmbh Pharmaceutical composition comprising a polymeric carrier cargo complex and at least one protein or peptide antigen
WO2014011705A1 (en) 2012-07-09 2014-01-16 The Regents Of The University Of California Vault immunotherapy
US20140234351A1 (en) 2013-02-14 2014-08-21 Immunocellular Therapeutics, Ltd. Cancer vaccines and vaccination methods
CA2905108C (en) 2013-03-14 2021-12-07 Julie HUGHES Cholestosome vesicles for incorporation of molecules into chylomicrons
US9163284B2 (en) 2013-08-09 2015-10-20 President And Fellows Of Harvard College Methods for identifying a target site of a Cas9 nuclease
AU2014310934B2 (en) 2013-08-21 2019-09-12 CureVac SE Respiratory syncytial virus (RSV) vaccine
US10369216B2 (en) 2014-04-01 2019-08-06 Curevac Ag Polymeric carrier cargo complex for use as an immunostimulating agent or as an adjuvant
EP3503876A4 (en) * 2016-08-23 2020-06-10 Jerome J. Schentag Cholesteryl ester vesicles loading peptides, proteins and nucleic acids into chylomicrons and body cells
US11898179B2 (en) 2017-03-09 2024-02-13 President And Fellows Of Harvard College Suppression of pain by gene editing
US20210196809A1 (en) * 2017-03-09 2021-07-01 President And Fellows Of Harvard College Cancer vaccine
WO2019139645A2 (en) 2017-08-30 2019-07-18 President And Fellows Of Harvard College High efficiency base editors comprising gam
DE112021002672T5 (en) 2020-05-08 2023-04-13 President And Fellows Of Harvard College METHODS AND COMPOSITIONS FOR EDIT BOTH STRANDS SIMULTANEOUSLY OF A DOUBLE STRANDED NUCLEOTIDE TARGET SEQUENCE

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5262177A (en) * 1986-02-07 1993-11-16 Oncogen Recombinant viruses encoding the human melanoma-associated antigen
US5342774A (en) * 1991-05-23 1994-08-30 Ludwig Institute For Cancer Research Nucleotide sequence encoding the tumor rejection antigen precursor, MAGE-1
US5994523A (en) * 1994-04-22 1999-11-30 The United States Of America As Represented By The Department Of Health And Human Services Melanoma antigens and their use in diagnostic and therapeutic methods
US6270778B1 (en) * 1994-04-22 2001-08-07 The United States Of America As Represented By The Department Of Health And Human Services Melanoma antigens and their use in diagnostic and therapeutic methods
US6500919B1 (en) * 1994-02-16 2002-12-31 Introgene B.V. Melanoma associated antigenic polypeptide, epitopes thereof and vaccines against melanoma
US6660276B1 (en) * 1994-02-16 2003-12-09 The University Of Virginia Patent Foundation Peptides recognized by melanoma-specific cytotoxic lymphocytes, and uses therefor

Family Cites Families (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS5990052A (en) * 1982-11-16 1984-05-24 Katsu Taniguchi Melanoma diagnosis medicine using monoclonal specific antibody
JPS6147500A (en) 1984-08-15 1986-03-07 Res Dev Corp Of Japan Chimera monoclonal antibody and its preparation
EP0173494A3 (en) 1984-08-27 1987-11-25 The Board Of Trustees Of The Leland Stanford Junior University Chimeric receptors by dna splicing and expression
GB8422238D0 (en) 1984-09-03 1984-10-10 Neuberger M S Chimeric proteins
JPS61134325A (en) 1984-12-04 1986-06-21 Teijin Ltd Expression of hybrid antibody gene
US5679511A (en) * 1986-10-06 1997-10-21 Donald Guthrie Foundation For Medical Research, Inc. CDNA clones for a regulatory protein in the melanin-production pathway
WO1990001443A1 (en) 1988-08-08 1990-02-22 Iffiu Michael A Collapsible recumbent bicycle with streamlined three-part folding canopy
CA2016841C (en) 1989-05-16 1999-09-21 William D. Huse A method for producing polymers having a preselected activity
AU2170692A (en) * 1991-06-05 1993-01-08 Bristol-Myers Squibb Company Me20: monoclonal antibodies and antigen for human melanoma
CA2128424A1 (en) * 1992-01-21 1993-07-22 Pierre G. Coulie Method for determining cytolytic t cell precursors
EP0563485A1 (en) 1992-03-30 1993-10-06 Schering-Plough In vitro generation of human dendritic cells and uses thereof
WO1993020185A1 (en) 1992-04-01 1993-10-14 Steinman Ralph M Method for in vitro proliferation of dendritic cell precursors and their use to produce immunogens
US6001975A (en) 1993-03-18 1999-12-14 Ludwig Institute For Cancer Research Precursor processed to at least one tumor rejection antigen presented by HLA-A2
US5620886A (en) * 1993-03-18 1997-04-15 Ludwig Institute For Cancer Research Isolated nucleic acid sequence coding for a tumor rejection antigen precursor processed to at least one tumor rejection antigen presented by HLA-A2
US5856091A (en) 1993-03-18 1999-01-05 Ludwig Institute For Cancer Research Isolated nucleic acid sequence coding for a tumor rejection antigen precursor processed to at least one tumor rejection antigen presented by HLA-A2
AU6496094A (en) * 1993-03-31 1994-10-24 Abbott Laboratories Tumor-associated antigens recognized by t cells and the uses of these antigens
US5837476A (en) 1995-03-03 1998-11-17 Ludwig Institute Methods for determining disorders by assaying for a non-tyrosinase, tumor rejection antigen precursor
US20040156861A1 (en) 1996-07-11 2004-08-12 Figdor Carl Gustav Melanoma associated peptide analogues and vaccines against melanoma

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5262177A (en) * 1986-02-07 1993-11-16 Oncogen Recombinant viruses encoding the human melanoma-associated antigen
US5342774A (en) * 1991-05-23 1994-08-30 Ludwig Institute For Cancer Research Nucleotide sequence encoding the tumor rejection antigen precursor, MAGE-1
US6500919B1 (en) * 1994-02-16 2002-12-31 Introgene B.V. Melanoma associated antigenic polypeptide, epitopes thereof and vaccines against melanoma
US6660276B1 (en) * 1994-02-16 2003-12-09 The University Of Virginia Patent Foundation Peptides recognized by melanoma-specific cytotoxic lymphocytes, and uses therefor
US5994523A (en) * 1994-04-22 1999-11-30 The United States Of America As Represented By The Department Of Health And Human Services Melanoma antigens and their use in diagnostic and therapeutic methods
US6270778B1 (en) * 1994-04-22 2001-08-07 The United States Of America As Represented By The Department Of Health And Human Services Melanoma antigens and their use in diagnostic and therapeutic methods

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8030280B2 (en) 1994-04-22 2011-10-04 The United States Of America As Represented By The Secretary, Department Of Health And Human Services Melanoma antigens and their use in diagnostic and therapeutic methods

Also Published As

Publication number Publication date
FI123050B (en) 2012-10-15
US8273724B2 (en) 2012-09-25
FI20115005A (en) 2011-01-04
US5874560A (en) 1999-02-23
US5844075A (en) 1998-12-01
US20110027299A1 (en) 2011-02-03
EP2311952A2 (en) 2011-04-20
CA2722364C (en) 2013-12-31
EP2311952A3 (en) 2011-06-01
US20110287043A1 (en) 2011-11-24
US5994523A (en) 1999-11-30
JP2010178739A (en) 2010-08-19
US8030280B2 (en) 2011-10-04
CA2722364A1 (en) 1995-11-02
US7803614B2 (en) 2010-09-28

Similar Documents

Publication Publication Date Title
US7803614B2 (en) Melanoma antigens and their use in diagnostic and therapeutic methods
US6965017B2 (en) Melanoma antigens and their use in diagnostic and therapeutic methods
EP0871725B1 (en) P15 melanoma antigens and their use in diagnostic and therapeutic methods
US7749967B2 (en) Peptides of a melanoma antigen and their use in diagnostic, prophylactic, and therapeutic methods

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: PAYOR NUMBER ASSIGNED (ORIGINAL EVENT CODE: ASPN); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STCF Information on status: patent grant

Free format text: PATENTED CASE

FPAY Fee payment

Year of fee payment: 4

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 8TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1552)

Year of fee payment: 8

FEPP Fee payment procedure

Free format text: MAINTENANCE FEE REMINDER MAILED (ORIGINAL EVENT CODE: REM.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

LAPS Lapse for failure to pay maintenance fees

Free format text: PATENT EXPIRED FOR FAILURE TO PAY MAINTENANCE FEES (ORIGINAL EVENT CODE: EXP.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20220928